US20030073149A1 - Antibody complexes and methods for immunolabeling - Google Patents

Antibody complexes and methods for immunolabeling Download PDF

Info

Publication number
US20030073149A1
US20030073149A1 US10/118,204 US11820402A US2003073149A1 US 20030073149 A1 US20030073149 A1 US 20030073149A1 US 11820402 A US11820402 A US 11820402A US 2003073149 A1 US2003073149 A1 US 2003073149A1
Authority
US
United States
Prior art keywords
labeling
antibody
protein
target
binding
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US10/118,204
Other versions
US8323903B2 (en
Inventor
Robert Archer
Joseph Beechem
David Hagen
Richard Haugland
Rosaria Haugland
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Life Technologies Corp
Original Assignee
Archer Robert A.
Beechem Joseph M.
Hagen David C.
Haugland Richard P.
Rosaria Haugland
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Archer Robert A., Beechem Joseph M., Hagen David C., Haugland Richard P., Rosaria Haugland filed Critical Archer Robert A.
Priority to US10/118,204 priority Critical patent/US8323903B2/en
Priority to EP02768949A priority patent/EP1442302B1/en
Priority to JP2003533851A priority patent/JP2006513398A/en
Priority to CA002462280A priority patent/CA2462280C/en
Priority to EP09163588A priority patent/EP2113773B1/en
Priority to AT02768949T priority patent/ATE436022T1/en
Priority to EP10181177A priority patent/EP2259067B1/en
Priority to PCT/US2002/031416 priority patent/WO2003030817A2/en
Priority to DE60232904T priority patent/DE60232904D1/en
Priority to GB0410515A priority patent/GB2397302B/en
Publication of US20030073149A1 publication Critical patent/US20030073149A1/en
Priority to US10/666,291 priority patent/US20070269902A1/en
Priority to JP2007083130A priority patent/JP4977512B2/en
Assigned to INVITROGEN CORPORATION reassignment INVITROGEN CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BEECHEM, JOSEPH M
Assigned to MOLECULAR PROBES, INC. reassignment MOLECULAR PROBES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HAGEN, DAVID C., BEECHEM, JOSEPH M., HAUGLAND, RICHARD P., HAUGLAND, ROSARIA P., ARCHER, ROBERT M.
Assigned to BANK OF AMERICA, N.A., AS COLLATERAL AGENT reassignment BANK OF AMERICA, N.A., AS COLLATERAL AGENT SECURITY AGREEMENT Assignors: Life Technologies Corporation
Assigned to Life Technologies Corporation reassignment Life Technologies Corporation ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MOLECULAR PROBES, INC.
Assigned to Life Technologies Corporation reassignment Life Technologies Corporation MERGER (SEE DOCUMENT FOR DETAILS). Assignors: INVITROGEN CORPORATION
Priority to US13/610,009 priority patent/US8535894B2/en
Publication of US8323903B2 publication Critical patent/US8323903B2/en
Application granted granted Critical
Assigned to Life Technologies Corporation reassignment Life Technologies Corporation LIEN RELEASE Assignors: BANK OF AMERICA, N.A.
Assigned to Life Technologies Corporation reassignment Life Technologies Corporation CORRECTIVE ASSIGNMENT TO CORRECT THE APPLICATION NO 09452626 PREVIOUSLY RECORDED ON REEL 023882 FRAME 0551. ASSIGNOR(S) HEREBY CONFIRMS THE MERGER SHOULD NOT HAVE BEEN RECORDED AGAINST THIS PATENT APPLICATION NUMBER. Assignors: INVITROGEN CORPORATION
Adjusted expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y30/00Nanotechnology for materials or surface science, e.g. nanocomposites
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y10/00Nanotechnology for information processing, storage or transmission, e.g. quantum computing or single electron logic
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6854Immunoglobulins
    • G01N33/6857Antibody fragments

Definitions

  • the present invention relates to novel immunolabeling compositions and methods for use in the detection and measurement of one or more targets in a biological sample.
  • the invention has applications in the fields of molecular biology, cell biology screening, immunohistochemistry, diagnostics, and therapeutics.
  • Immunolabeling is a method for qualitative or quantitative determination of the presence of a target in a sample, wherein antibodies are utilized for their specific binding capacity.
  • the antibodies form a complex with the target (antigen), wherein a detectable label is present on the antibody or on a secondary antibody.
  • the detectable label is a key feature of immunolabeling, which can be detected directly or indirectly.
  • the label provides a measurable signal by which the binding reaction is monitored providing a qualitative and/or quantitative measure of the degree of binding.
  • the relative quantity and location of signal generated by the labeled antibodies can serve to indicate the location and/or concentration of the target.
  • the label can also be used to select and isolate labeled targets, such as by flow sorting or using magnetic separation media.
  • labels include but are not limited to radioactive nucleotides ( 125 I, 3 H, 14 C, 32 P), chemiluminescent, fluorescent, or phosphorescent compounds (e.g., dioxetanes, xanthene, or carbocyanine dyes, lanthanide chelates), particles (e.g., gold clusters, colloidal gold, microspheres, quantum dots), and enzymes (e.g., peroxidases, glycosidases, phosphatases, kinases).
  • the label is attached to the antibody in a manner that does not perturb the antibody's binding characteristics but enables the label to be measured by an appropriate detection technology.
  • the choice of labels is influenced by factors such as ease and sensitivity of detection, equipment availability, background in the sample (including other labels) and the degree to which such labels are readily attached to the particular antibody.
  • Both direct and indirect labeling of antibodies are utilized for immunolabeling.
  • Direct labeling utilizes only a primary antibody, i.e. the antibody specific for the target, bound to the label.
  • indirect labeling utilizes a secondary antibody bound to the label, which is specific for the primary antibody, e.g. a goat anti-rabbit antibody.
  • the principal differences in immunolabeling methods and materials reside in the way that the label is attached to the antibody-antigen complex, the type of label that is used, and the means by which the antibody-antigen complex is detected.
  • Limitations for direct labeling primary antibodies include the need for buffers free of primary amines, or carrier proteins such as bovine serum albumin (BSA), and other compounds such as tris-(hydroxymethyl)aminomethane (TRIS), glycine, and ammonium ions. These materials are, however, common components in antibody buffers and purification methods, and it may not be possible or feasible to remove them prior to the coupling reaction. In particular, many monoclonal antibodies are available only as ascites fluid or in hybridoma culture supernatants, or diluted with carrier proteins, such as albumins. Thus, direct labeling of antibodies in ascites fluid or other medias containing interfering compounds is not attainable.
  • the indirect immunolabeling method typically involves a multi-step process in which an unlabeled first antibody (typically a primary antibody) is directly added to the sample to form a complex with the antigen in the sample. Subsequently, a labeled secondary antibody, specific for the primary antibody, is added to the sample, where it attaches noncovalently to the primary antibody-antigen complex.
  • a detectable label is covalently attached to an immunoglobulin-binding protein such as protein A and protein G to detect the antibody-antigen complex that has previously been formed with the target in the sample.
  • ligands such as streptavidin, that are meant to amplify the detectable signal also expands this cascade binding.
  • Indirect immunolabeling often results in false positives and high background. This is due to the fact that secondary antibodies, even when purified by adsorption against related species, nevertheless can exhibit significant residual cross-reactivity when used in the same sample. For example, when mouse tissue is probed with a mouse monoclonal antibody, the secondary antibody must necessarily be a labeled anti-mouse antibody. This anti-mouse antibody will detect the antibody of interest but will inevitably and additionally detect irrelevant, endogenous mouse immunoglobulins inherent in mouse tissue. This causes a significant background problem, especially in diseased tissues, which reduces the usefulness and sensitivity of the assay.
  • the simultaneous detection of more than one primary antibody in a sample without this significant background interference depends on the availability of secondary antibodies that 1) do not cross-react with proteins intrinsic to the sample being examined, 2) recognize only one of the primary antibodies, and 3) do not recognize each other (Brelje, et al., METHODS IN CELL BIOLOGY 38,97-181, especially 111-118 (1993)).
  • Another strategy for blocking access to endogenous immunoglobulins in the sample involves pre-incubating the sample with a monovalent antibody, such as Fab′ fragments, from an irrelevant species that recognize endogenous immunoglobulins.
  • Fab′ fragments monovalent antibody
  • This approach requires large quantities of expensive Fab′ fragments and gives mixed results and adds at least two steps (block and wash) to the overall staining procedure.
  • the addition of a cross-linking reagent has resulted in improved reduction of background levels (Tsao, et al., U.S. Pat. No. 5,869,274 (1997)) but this is problematic when used with fluorophore-labeled antibodies.
  • the cross-linking causes an increase in the levels of autofluorescence (J. Neurosci. Meth.
  • a multi-step sequential-labeling procedure is used to overcome the problems of cross-reactivity.
  • the sample is incubated with a first antibody to form a complex with the first antigen, followed by incubation of the sample with a fluorophore-labeled goat Fab anti-mouse IgG to label the first antibody and block it from subsequently complexing when the second antibody is added.
  • a second mouse antibody forms a complex with the second antigen. Being blocked from cross-reacting with the first antibody, the second mouse antibody is detected with a standard indirect-labeling method using a goat anti-mouse antibody conjugated to a different fluorescent dye (J. Histochem. Cytochem. 34, 703 (1986)).
  • This process is complex in that it requires multiple incubation steps and washing steps and it still cannot be used with mouse antibodies to probe mouse tissue.
  • Another blocking method is disclosed in the animal research kit (ARK) developed by DAKO.
  • ARK animal research kit
  • a primary antibody is complexed with biotin-labeled goat Fab anti-mouse IgG and excess free Fab is blocked with normal mouse serum.
  • the Fab used in this process is generated from the intact IgG (rather than a selected region) there is a potential for the formation of anti-paratope or anti-idiotype antibodies that will block the antigen-binding site and prevent immunolabeling.
  • the biotinylated antibody also requires subsequent addition of a labeled avidin or streptavidin conjugate for its subsequent visualization.
  • the present invention is advantageous over previously described methods and compositions in that it provides the benefits of indirect labeling with the easy and flexibility of direct labeling for determination of a desired target in a biological sample.
  • the present invention provides labeled monovalent proteins specific for a target-binding antibody, which are complexed prior to addition with a biological sample. Because these monovalent proteins are not bivalent antibodies, precipitation and cross-linking are not a problem. Therefore the compositions of the present invention can be used with immunologically similar monoclonal or polyclonal antibodies of either an identical isotype or different isotypes.
  • the monovalent labeling proteins are specific for the Fc region of target-binding antibodies, these proteins will not interfere with the binding region of the primary antibody.
  • the monovalent labeling proteins are not negatively affected by the presence of primary amines like BSA, gelatin, hybridoma culture supernatants or ascites fluid, thus primary antibodies present in these media can be effectively labeled with the labeling proteins of the present invention.
  • the present invention provides numerous advantages over the conventional methods of immunolabeling.
  • Methods and compositions are provided for determining the presence, absence or location of a desired target in a biological sample.
  • the methods initially involve pre-forming the immunolabeling complex, the target-binding antibody and the labeling protein, followed by addition to a biological sample and determination of the desired target.
  • the methods of the invention are broadly applicable to immunolabeling any antigen target in any sample from any origin.
  • FIG. 1 is a schematic representation of the formation of the immunolabeling complex (target-bindingprotein and labeling protein).
  • FIG. 2 graphically represents species specificity of goat Fab anti-(mouse Fc), as observed using a microplate coated with IgG of various species.
  • the various species were blocked with BSA, reacted with biotinylated goat Fab anti-(mouse Fc), washed, and then treated with streptavidin-horseradish peroxidase (HRP), followed by hydrogen peroxide (H 2 O 2 ) and the Amplex Red peroxidase detection reagent.
  • HRP streptavidin-horseradish peroxidase
  • FIG. 3 graphically depicts the optimal molar ratio of a goat Fab anti-(mouse Fc) protein. Varying amounts of an Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse Fc) were added to a constant amount of anti-biotin monoclonal antibody (mAb). This mixture was equilibrated for 20 minutes, and then added to biotinylated-BSA in a microplate well. After allowing time to bind, the plates were washed and the remaining fluorescence was quantitated. The analysis was performed in triplicate (circles). Control experiments were performed, as described above, but without adding the primary anti-biotin antibody (solid squares).
  • mAb anti-biotin monoclonal antibody
  • FIG. 4 illustrates a comparison of the fluorescence intensity (Example 6) for labeling protein prepared in homogeneous solution (Example 4) and labeling protein prepared on a column (Example 5).
  • FIG. 5A CD3-positive R-phycoerythrin (R-PE) stained T cells are shown in the upper left (UL) and upper right (UR) quadrants (Example 18).
  • the relative percentages of total lymphocytes that are CD3-positive cells are 83.3% (UL+UR).
  • the relative percentage of CD8-positive Alexa Fluor 488 dye-stained lymphocytes and CD3-positive R-PE dye-stained lymphocytes is 35.1% (UR quadrant).
  • the lower left quadrant shows CD3-negative lymphocytes (i.e. non-T cells) comprised of NK cells, B cells and some monocytes. In the lower right (LR, 2.7%) region are non-T cells, which are nonspecifically stained.
  • FIG. 5B CD3-positive T-cells are subdivided into Alexa Fluor 647 CD4-positive and Alexa Fluor 488 CD8-positive (Example 18).
  • CD4-positive cells represent 50.9% of total lymphocytes (UL quadrant) and CD8-positive cells represent 24.5% of the total lymphocytes (LR quadrant).
  • the 23.1% of cells in the LL quadrant are non-T cells, while the 1.5% of cells in UR quadrant are likely nonspecifically stained lymphocytes.
  • FIG. 6 illustrates high-performance size-exclusion chromatographic analysis of Alexa Fluor 488 dye-labeled goat Fab anti-(mouse Fc) labeling protein binding to a mouse IgG 1 target-binding antibody.
  • the labeling protein alone, appears as a peak at 38 minutes; the target-binding antibody, alone, appears as a peak at 33 minutes.
  • labeling protein and target-binding antibody are mixed together at a molar ratio of ⁇ 5:1 (labeling protein:target-binding antibody), the resulting immunolabeling complex appears as a peak at 29 minutes (Example 10).
  • FIG. 7 is a cartoon depiction of the process for preparing improved labeling proteins.
  • “Affinity” is defined as the strength of the binding interaction of two molecules, such as an antigen and its antibody, which is defined for antibodies and other molecules with more than one binding site as the strength of binding of the ligand at one specified binding site.
  • “High affinity” is for a ligand that binds to an antibody having an affinity constant (K a ) of greater than 10 4 M ⁇ 1 , typically 10 5 -10 11 M ⁇ 1 ; as determined by inhibition ELISA or an equivalent affinity determined by comparable techniques such as, for example, Scatchard plots or using K d /dissociation constant, which is the reciprocal of the K a , etc.
  • Antibody is a protein of the immunoglobulin (Ig) superfamily that binds noncovalently to certain substances (e.g. antigens and immunogens) to form an antibody-antigen complex, including but not limited to antibodies produced by hybridoma cell lines, by immunization to elicit a polyclonal antibody response, by chemical synthesis, and by recombinant host cells that have been transformed with an expression vector that encodes the antibody.
  • the immunoglobulin antibodies are classified as IgA, IgD, IgE, IgG, and IgM and members of each class are said to have the same isotype.
  • Human IgA and IgG isotypes are further subdivided into subtypes IgA 1 , and IgA 2 , and IgG 1 , IgG 2 , IgG 3 , and IgG 4 .
  • Mice have generally the same isotypes as humans, but the IgG isotype is subdivided into IgG 1 , IgG 2a , IgG 2b , and IgG 3 subtypes.
  • antibody as used herein includes within its scope (a) any of the various classes or sub-classes of immunoglobulin, e.g., IgG, IgM, IgE derived from any of the animals conventionally used and (b) polyclonal and monoclonal antibodies, such as murine, chimeric, or humanized antibodies.
  • Antibody molecules have regions of amino acid sequences that can act as an antigenic determinant, e.g. the Fc region, the kappa light chain, the lambda light chain, the hinge region, etc.
  • An antibody that is generated against a selected region is designated anti-[region], e.g.
  • antibody is typically generated against an antigen by immunizing an organism with a macromolecule to initiate lymphocyte activation to express the immunoglobulin protein.
  • the term antibody also covers any polypeptide or protein having a binding domain that is, or is homologous to, an antibody binding domain, including, without limitation, single-chain Fv molecules (scFv), wherein a VH domain and a VL domain are linked by a peptide linker that allows the two domains to associate to form an antigen binding site (Bird et al., Science 242, 423 (1988) and Huston et al., Proc. Natl. Acad. Sci. USA 85, 5879 (1988)). These can be derived from natural sources, or they may be partly or wholly synthetically produced.
  • Antibody fragments refers to fragments of antibodies that retain the principal selective binding characteristics of the whole antibody. Particular fragments are well-known in the art, for example, Fab, Fab′, and F(ab′) 2 , which are obtained by digestion with various proteases and which lack the Fc fragment of an intact antibody or the so-called “half-molecule” fragments obtained by reductive cleavage of the disulfide bonds connecting the heavy chain components in the intact antibody. Such fragments also include isolated fragments consisting of the light-chain-variable region, “Fv” fragments consisting of the variable regions of the heavy and light chains, and recombinant single chain polypeptide molecules in which light and heavy variable regions are connected by a peptide linker.
  • binding fragments include (i) the Fd fragment, consisting of the VH and CH1 domains; (ii) the dAb fragment (Ward, et al., Nature 341, 544 (1989)), which consists of a VH domain; (iii) isolated CDR regions; and (iv) single-chain Fv molecules (scFv) described above.
  • arbitrary fragments can be made using recombinant technology that retains antigen-recognition characteristics.
  • Antigen means a molecule that induces, or is capable of inducing, the formation of an antibody or to which an antibody binds selectively, including but not limited to a biological material. Antigen also refers to “immunogen”. An antibody binds selectively to an antigen when there is a relative lack of cross-reactivity with or interference by other substances present.
  • Bio material means a material of biological origin or a synthetic variant of a material of biological origin, including but not limited to cells, bodily fluids, membranes, proteins, amino acids, nucleic acids, carbohydrates, lipids, and any polymers or combinations thereof, including natural as well as synthetic polymers that are optionally relatively homogeneous or heterogeneous.
  • Biological Sample refers to any material that may contain a target to which an antibody can bind.
  • the sample comprises tissue, cell or cells, cell extracts, cell homogenates, purified or reconstituted proteins, recombinant proteins, bodily and other biological fluids, viruses or viral particles, prions, subcellular components, or synthesized proteins.
  • Possible sources of cellular material used to prepare the sample of the invention include without limitation plants, animals, fungi, protists, bacteria, archae, or cell lines derived from such organisms.
  • Biotin means any biotin derivative, including without limitation, substituted and unsubstituted biotin, and analogs and derivatives thereof, as well as substituted and unsubstituted derivatives of caproylamidobiotin, biocytin, desthiobiotin, desthiobiocytin, iminobiotin, and biotin sulfone.
  • Biotin-binding protein means any protein that binds selectively and with high affinity to biotin, including without limitation, substituted or unsubstituted avidin, and analogs and derivatives thereof, as well as substituted and unsubstituted derivatives of streptavidin, ferritin avidin, nitroavidin, nitrostreptavidin, and NeutravidinTM avidin (a de-glycosylated modified avidin having an isoelectric point near neutral).
  • Body fluid means a fluid substance from the body, including but not limited to, for example, circulating blood and lymph, the chyle, the gastric, pancreatic, and intestinal juices, the saliva, bile, urine, aqueous humor, spinal fluid, muscle serum and ascites.
  • Buffer means a system that acts to minimize the change in concentration of a specific chemical species in solution against addition or depletion of this species.
  • Cell or cells means an autonomous self-replicating unit composed of protoplasm delimited by a cell membrane that may constitute an organism (in the case of unicellular organisms) or be a subunit of multicellular organisms (in which individual cells may be more or less specialized differentiated) for particular functions or plant cells.
  • Cell or cells as used in the instant application also includes cells in cell culture medium, single cells, isolated cells, single-cell organisms; and “portions thereof” including, but not limited to, cell extracts, cell homogenates, cell lysates, and subcellular components.
  • “Complex” means two or more molecules held together by noncovalent bonding, which are typically noncovalent combinations of biomolecules such as a protein complexed with another protein.
  • a protein is covalently labeled with a substance when there is a covalent chemical bond between the substance and the protein.
  • Detectable response means a change in, or occurrence of, a signal that is detectable either by observation or instrumentally.
  • the detectable response is an optical response resulting in a change in the wavelength distribution patterns or intensity of absorbance or fluorescence or a change in light scatter, fluorescence lifetime, fluorescence polarization, or a combination of the above parameters.
  • Other detectable responses include, for example, chemiluminescence, phosphorescence, radiation from radioisotopes, magnetic attraction, and electron density.
  • Detectably distinct means the signal is distinguishable or separable by a physical property either by observation or instrumentally.
  • a fluorophore is readily distinguishable, either by spectral characteristics or by fluorescence intensity, lifetime, polarization or photo-bleaching rate from another fluorophore in the sample, as well as from additional materials that are optionally present.
  • Directly detectable means that the presence of a material or the signal generated from the material is immediately detectable by observation, instrumentation, or film without requiring chemical modifications or additional substances.
  • drug-labeled protein means a labeling protein of the invention, where instead of a detectable label being attached to the protein, a therapeutic agent or drug is attached.
  • drug-labeled protein is used interchangeably with immunoconjugate.
  • Enzyme means a protein molecule produced by living organisms, or through chemical modification of a natural protein molecule, that catalyses chemical reactions of other substances without itself being destroyed or altered upon completion of the reactions.
  • other substances include but are not limited to, chemiluminescent, chromogenic, and fluorogenic substances.
  • Immunoconjugates are labeling proteins of the invention, where instead of a detectable label being attached to the protein, a therapeutic agent or drug is attached.
  • the term immunoconjugate is used interchangeably with drug-labeled protein.
  • Immunolabeling complex means an antibody-protein complex made detectable, traceable, or therapeutically useful by incorporation of a detectable label, drug, or therapeutic into the protein that is noncovalently and selectively bound to the antibody.
  • Kit means a packaged set of related components, typically one or more compounds or compositions.
  • Label means a chemical used to facilitate identification and/or quantitation of a target substance.
  • Illustrative labels include labels that can be directly observed or measured or indirectly observed or measured.
  • Such labels include, but are not limited to, radiolabels that can be measured with radiation-counting devices; pigments, dyes or other chromogens that can be visually observed or measured with a spectrophotometer; spin labels that can be measured with a spin label analyzer; and fluorescent moieties, where the output signal is generated by the excitation of a suitable molecular adduct and that can be visualized by excitation with light that is absorbed by the dye or can be measured with standard fluorometers or imaging systems, for example.
  • the label can be a luminescent substance such as a phosphor or fluorogen; a bioluminescent substance; a chemiluminescent substance, where the output signal is generated by chemical modification of the signal compound; a metal-containing substance; or an enzyme, where there occurs an enzyme-dependent secondary generation of signal, such as the formation of a colored product from a colorless substrate.
  • the label may also take the form of a chemical or biochemical, or an inert particle, including but not limited to colloidal gold, microspheres, quantum dots, or inorganic crystals such as nanocrystals or phosphors (see, e.g., Beverloo, et al., Anal. Biochem. 203, 326-34 (1992)).
  • label can also refer to a “tag” or hapten that can bind selectively to a labeled molecule such that the labeled molecule, when added subsequently, is used to generate a detectable signal.
  • a tag or hapten that can bind selectively to a labeled molecule such that the labeled molecule, when added subsequently, is used to generate a detectable signal.
  • HRP horseradish peroxidase
  • a chromogenic substrate e.g., tetramethylbenzidine
  • fluorogenic substrate such as Amplex Red or Amplex Gold (Molecular Probes, Inc.
  • the tag can be a hapten or antigen (e.g., digoxigenin), and an enzymatically, fluorescently, or radioactively labeled antibody can be used to bind to the tag.
  • hapten or antigen e.g., digoxigenin
  • an enzymatically, fluorescently, or radioactively labeled antibody can be used to bind to the tag.
  • labels include, but are not limited to, particles, fluorescent dyes, haptens, enzymes and their chromogenic, fluorogenic, and chemiluminescent substrates, and other labels that are described in the MOLECULAR PROBES HANDBOOK OF FLUORESCENT PROBES AND RESEARCH CHEMICALS by Richard P. Haugland, 6 th Ed., (1996), and its subsequent 7 th edition and 8 th edition updates issued on CD Rom in November 1999 and May 2001, respectively, the contents of which are incorporated by reference, and in other published sources.
  • “Monovalent antibody fragment” refers to an antibody fragment that has only one antigen-binding site.
  • monovalent antibody fragments include, but are not limited to, Fab fragments (no hinge region), Fab′ fragments (monovalent fragments that contain a heavy chain hinge region), and single-chain fragment variable (ScFv) proteins.
  • Non-antibody immunoglobulin-binding protein means a protein that binds selectively and noncovalently to a member of the Ig superfamily of proteins, including but not limited to proteins A, G, and L, hybrids thereof (A/G), recombinant versions and cloned versions thereof, fusions of these proteins with detectable protein labels, and lectins but the protein itself is not an antibody or an antibody fragment.
  • Non-antibody immunoglobulin-binding peptide means a peptide that selectively and noncovalently binds to a member of the Ig superfamily of proteins, such as a peptide that is selected by a process of screening against a biomolecular library.
  • Peptide means a polymer of amino acids, and includes polypeptides.
  • Protein G means proteins comprising one or more natural IgG-binding domains of protein G, hybrids of the natural IgG-binding domains, and mutants thereof wherein the variant retains the capability of binding IgG, or fragments thereof.
  • Protein A means proteins comprising one or more natural IgG-binding domains of protein A, hybrids of the natural IgG-binding domains, and mutants thereof wherein the variant retains the capability of binding IgG, or fragments thereof.
  • Protein L means proteins comprising one or more natural antibody light-chain-binding domains of protein L, hybrids of the natural antibody light-chain-binding domains, and mutants thereof wherein the variant retains the capability of binding antibody light chain, or fragments thereof.
  • “Selectively binds” refers to the situation in which one member of a specific intra or inter species binding pair will not show any significant binding to molecules other than its specific intra- or inter-species binding partner (e.g., an affinity of about 100-fold less), i.e. minimal cross-reactivity.
  • Subcellular component means functional units within a cell, including but not limited to, for example, membranes, organelles, receptors, nucleus, chromosomes, chloroplasts, peroxisomes, polyribosomes, mitochondria, lysosomes, DNA, RNA, endogenous proteins, peptides, enzymes, hormones, and factors.
  • Target means any entity to be detected by the association of the target-binding antibody with it.
  • the immunolabeling complex which contains the target-binding antibody and the labeling protein bound by a detectable label, is pre-formed followed by the addition to a sample suspected of containing the desired target.
  • the labeling protein is either a monovalent Fab, fragment or a non-immunoglobulin peptide or protein, and specifically binds a selected region of the target-binding antibody.
  • the labeling protein is covalently attached to one or more detectable labels, wherein the detectable labels can be the same or different allowing for multiparameter applications.
  • the labeling proteins bind selectively and with high affinity to a selected region of the target-binding antibody. These labeling proteins bind selectively to the same region of the target-binding antibody.
  • the binding region for the labeling proteins may be a selected peptide linker (including the J region), light chain or heavy chain of the target-binding antibody; preferably the labeling protein binds the Fc region of the target-binding antibody.
  • the target-binding antibody may be bound with one or more labeling proteins and the labeling proteins may be bound with one label, two similar labels, two distinct labels or multiple combinations thereof.
  • Binding of more than one label permits combinatorial applications that increase the number of detectable targets in the sample.
  • the same dye is used on a given labeling protein and the labeling proteins are monovalent Fab fragments or non-antibody peptide or proteins.
  • the labeling protein can be either an antibody fragment, such as, but not limited to, anti-Fc, an anti-Fc isotype, anti-J chain, anti-kappa light chain, anti-lambda light chain, or a single-chain fragment variable protein; or a non-antibody peptide or protein, such as, for example but not limited to, soluble Fc receptor, protein G, protein A, protein L, lectins, or a fragment thereof.
  • the labeling protein is a Fab fragment specific to the Fc portion of the target-binding antibody or to an isotype of the Fc portion of the target-binding antibody.
  • the monovalent Fab fragments are produced from either murine monoclonal antibodies or polyclonal antibodies generated in a variety of animals, for example but not limited to, rabbit or goat. These fragments can be generated from any isotype such as murine IgM, IgG 1 , IgG 2a , IgG 2b or IgG 3 .
  • a non-antibody protein or peptide such as protein G, or other suitable proteins, can be used alone or coupled with albumin wherein albumin is complexed with the detectable label.
  • Preferred albumins of the invention include human and bovine serum albumins or ovalbumin.
  • Labels that are covalently attached to the labeling proteins are known by those of skill in the art and include, but are not limited to, radiolabels, pigments, dyes or other chromogens, spin labels, fluorescent compounds, haptens, electron transfer agents, and particles.
  • the label can also be a precursor to a luminescent substance, a bioluminescent substance, a chemiluminescent substance, or a metal-containing substance.
  • Preferred labels are fluorescent moieties such as those described in the MOLECULAR PROBES HANDBOOK OF FLUORESCENT PROBES AND RESEARCH CHEMICALS by R. P. Haugland 8 th Ed., CD-ROM (2001).
  • xanthenes e.g., fluoreceins, rhodamines and rhodols
  • sulfonated xanthenes such as those disclosed in U.S. Pat. No. 6,130,101 and UK 9611997.9 and fluorinated xanthenes such as those disclosed in U.S. Pat. No. 6,162,931
  • coumarins especially sulfonated coumarins such as those described in U.S. Pat. No. 5,969,157 and fluorinated coumarins such as those disclosed in U.S. Pat. No.
  • cyanines especially sulfonated cyanines disclosed and PCT/01/30404 publication.
  • Other preferred labels are fluorescent proteins, especially phycobiliprotein and tandem-dye conjugates thereof as well as haptens such as biotin including desthibiotin) and enzymes, described in greater detail below.
  • a particular advantage of the current invention over other methods of labeling target-binding antibodies is that it provides for a novel method to directly label the antibodies in any solution containing primary amines or non-antibody proteins, for example from ascites fluid, hybridoma culture supernatant, serum, in the presence of stabilizing proteins like BSA, gelatin, ammonium ions, or buffers like TRIS. Consequently, the methods of the instant invention provide a means to rapidly screen hybridomas for antibody expression and quality of antibody properties (e.g. affinity, specificity, cross-reactivity, isotype, abundance).
  • the labeling complexes are combined with a sample thought to contain target materials in any way that facilitates contact between the immunolabeling complexes and the target of interest.
  • Preferred targets of the invention are biological materials and include, for example, but not limited to, a cell, receptor, membrane, protein, nucleic acid, or carbohydrate, but can be any antigenic determinant.
  • the sample Prior to combination with the immunolabeling complexes, the sample is prepared in a way that makes the target materials in the sample accessible to the immunolabeling complexes.
  • the samples used in the invention are comprised of tissue, cells, cell extracts, cell homogenates, purified or reconstituted proteins, recombinant proteins, biological fluids, or synthesized proteins.
  • Large macromolecules such as immunolabeling complexes tend to be impermeant to membranes of live biological cells. Treatments that permeabilize the plasma membrane, such as electroporation, shock treatments, or high extracellular ATP, can be used to introduce the labeling mixture and its immunolabeling complexes into cells.
  • the labeling mixture and its immunolabeling complexes can be physically inserted into cells, e.g. by pressure microinjection, scrape loading, patch-clamp methods, or phagocytosis.
  • the target materials may require purification or separation prior to addition of the immunolabelling complexes, which will depend on the way the antigenic determinants are contained in the sample.
  • the purified target materials may still be mixtures of different materials.
  • purified protein or nucleic acid mixtures may contain several different proteins or nucleic acids.
  • the purified target materials may be electrophoresed on gels such as agarose or polyacrylamide gels to provide individual species of target materials that may be subsequently blotted onto a polymeric membrane or detected within the gel matrix.
  • Preparation of a sample containing purified nucleic acids or proteins generally includes denaturation and neutralization.
  • DNA may be denatured by incubation with base (such as sodium hydroxide) or heat.
  • RNA is also denatured by heating (for dot blots) or by electrophoresing in the presence of denaturants such as urea, glyoxal, or formaldehyde, rather than through exposure to base (for Northern blots).
  • Proteins are denatured by heating in combination with incubation or electrophoresis in the presence of detergents such as sodium dodecyl sulfate.
  • the nucleic acids are then neutralized by the addition of an acid (e.g., hydrochloric acid), chilling, or addition of buffer (e.g., Tris, phosphate or citrate buffer), as appropriate.
  • an acid e.g., hydrochloric acid
  • buffer e.g., Tris, phosphate or citrate buffer
  • the preparation of a sample containing purified target materials further comprises immobilization of the target materials on a solid or semi-solid support.
  • Purified nucleic acids are generally spotted onto filter membranes such as nitrocellulose filters or nylon membranes in the presence of appropriate salts (such as sodium chloride or ammonium acetate) for DNA spot blots.
  • the purified nucleic acids are transferred to nitrocellulose filters by capillary blotting or electroblotting under appropriate buffer conditions (for Northern or Southern blots).
  • standard cross-linking techniques are used (for example, nitrocellulose filters are baked at 80° C. in vacuum; nylon membranes are subjected to illumination with 360 nm light).
  • the filter membranes are then incubated with solutions designed to prevent nonspecific binding of the nucleic acid probe (such as BSA, casein hydrolysate, single-stranded nucleic acids from a species not related to the probe, etc.) and hybridized to probes in a similar solution.
  • the nucleic acid probe such as BSA, casein hydrolysate, single-stranded nucleic acids from a species not related to the probe, etc.
  • Purified proteins are generally spotted onto nitrocellulose or nylon filter membranes after heat and/or detergent denaturation. Alternatively, the purified proteins are transferred to filter membranes by capillary blotting or electroblotting under appropriate buffer conditions (for Western blots).
  • Nonspecifically bound probe is washed from the filters with a solution such as saline-citrate or phosphate buffer. Filters are again blocked, to prevent nonspecific adherence of immunolabeling complexes. Finally, samples are mixed with immunolabeling complexes. Nonspecifically bound immunolabeling complex
  • the sample contains cellular nucleic acids (such as chromosomal or plasmid-borne genes within cells, RNA or DNA viruses or mycoplasma infecting cells, or intracellular RNA) or proteins
  • preparation of the sample involves lysing or permeabilizing the cell, in addition to the denaturation and neutralization already described.
  • Cells are lysed by exposure to agents such as detergent (for example sodium dodecyl sulfate, Tween, sarkosyl, or Triton), lysozyme, base (for example sodium, lithium, or potassium hydroxide), chloroform, or heat.
  • agents such as detergent (for example sodium dodecyl sulfate, Tween, sarkosyl, or Triton), lysozyme, base (for example sodium, lithium, or potassium hydroxide), chloroform, or heat.
  • Cells are permeabilized by conventional methods, such as by formaldehyde in buffer.
  • preparation of the sample containing cellular target materials typically further comprises immobilization of the target materials on a surface such as a solid or semi-solid support.
  • the targets may be arrayed on the support in a regular pattern or randomly.
  • These supports include such materials as slides, beads, optical fibers, and membranes.
  • the beads are preferably fluorescent or nonfluorescent polystyrene
  • the slides and optical fibers are preferably glass or plastic
  • the membrane is preferably poly(vinylidene difluoride) or nitrocellulose.
  • Permeabilized cells are typically fixed on microscope slides with known techniques used for in situ hybridization and hybridization to chromosome “squashes” and “spreads,” (e.g., with a reagent such as formaldehyde in a buffered solution). Alternatively, the samples used may be in a gel or solution.
  • unbound immunolabeling complexes that do not bind to the target are optionally removed from the sample by conventional methods, such as washing.
  • the bound immunolabeling complex that binds to the target can be fixed in place with the usual fixatives (e.g. formaldehyde, glutaraldehyde) and fixation methods. Fixation can be utilized to improve the durability of the sample and to prevent transfer of the noncovalently complexed labeling protein to other targeting antibodies in the sample that have the same specific binding region.
  • Detection of the bound label is performed using methods and reagents well known to those skilled in the art.
  • a preferred method of detection of the invention is through the use of fluorescence. Fluorescence from the immunolabeling complex binding to the target can be visualized with a variety of imaging techniques, including ordinary light or fluorescence microscopy, confocal laser-scanning microscopy, and flow cytometry, optionally using image deconvolution algorithms. Three-dimensional imaging resolution techniques in confocal microscopy utilize knowledge of the microscope's point spread function (image of a point source) to place out-of-focus light in its proper perspective.
  • Multiple-labeled target materials are optionally resolved spatially, chronologically, by size, or using detectably different spectral characteristics (including excitation and emission maxima, fluorescence intensity, fluorescence lifetime, fluorescence polarization, fluorescence photobleaching rates, or combinations thereof), or by combinations of these attributes.
  • multiple-labeled target materials are resolved using different labeling proteins with distinct spectral characteristics for each target material.
  • the labels on the labeling proteins are the same but the samples are labeled and viewed sequentially or are spatially separated.
  • enzymes can be used where there occurs an enzyme-dependent secondary generation of signal, such as the formation of a colored product from a non-colored substrate.
  • Enzyme labels or enzyme labeling systems are desirable in that they can achieve signal amplification and greater distinctions from backgrounds.
  • the enzyme breaks down a substrate to produce a chromophore or fluorophore or other detectable signal, thus amplifying the sensitivity of the assay and, if the substrate yields a distinct product at or near its site of formation, visualizing the site of the antigen/antibody complex in the sample.
  • the substrate is selected to yield the preferred measurable product. Chromogenic, fluorogenic and chemiluminescence-generating enzyme substrates are preferred.
  • enzymes are enzymes for which substrates yielding useful chromophores, fluorophores, or chemiluminescence are known. Such substrates are extensively used in the art and are described the MOLECULAR PROBES HANDBOOK OF FLUORESCENT PROBES AND RESEARCH CHEMICALS by R. P. Haugland 6 th Ed., (1996) and its subsequent 7 th edition and 8 th edition updates issued on CD Rom in November 1999 and May 2001, respectively, the contents of which are incorporated by reference, and in other published sources.
  • Preferred enzyme substrates of the invention are enzyme substrates that yield a fluorescent product that localizes at or near the site of enzyme activity.
  • Enzymes of use in the method include any enzymes that utilize a chromogenic, fluorogenic, or chemiluminescence-generating substrate.
  • Preferred enzymes of the invention include peroxidases, phosphatases, glycosidases, aequorins, or luciferases, and more specifically, HRP, Coprinus cinereus peroxidase, Arthromyces ramosus peroxidase, alkaline phosphatase, ⁇ -galactosidase, ⁇ -glucuronidase, or a protein A or protein G fusion protein of luciferase.
  • a preferred chromogenic (and in some cases fluorogenic) substrate and enzyme combination uses oxidoreductases such as horseradish peroxidase, Coprinus cinereus peroxidase, or Arthromyces ramosus peroxidase and a substrate such as 3,3′-diaminobenzidine (DAB) or 3-amino-9-ethylcarbazole (AEC), which yield a distinguishing color (brown and red, respectively).
  • DAB 3,3′-diaminobenzidine
  • AEC 3-amino-9-ethylcarbazole
  • chromogenic oxidoreductase substrates that yield detectable products include, but are not limited to: 2,2′-azino-bis(3-ethylbenzthiazoline-6-sulfonic acid (ABTS), o-phenylenediamine (OPD), 3,3′,5,5′-tetramethylbenzidine (TMB), o-dianisidine, 5-aminosalicylic acid, and 4-chloro-1-naphthol.
  • Fluorogenic substrates include, but are not limited to, homovanillic acid or 4-hydroxy-3-methoxyphenylacetic acid, reduced phenoxazines and reduced benzothiazines, including the Amplex Red reagent and its variants (Miike, U.S. Pat.
  • Peroxidase substrates that are tyramides as described in U.S. Pat. Nos. 5,196,306; 5,583,001 and 5,731,158, which are incorporated by reference, represent a unique class of peroxidase substrates in that they can be intrinsically detectable before action of the enzyme but are “fixed in place” by the action of a peroxidase in the process termed tyramide signal amplification (TSA).
  • TSA tyramide signal amplification
  • Another preferred chromogenic (and in some cases fluorogenic) substrate and enzyme combination uses a phosphatase enzyme such as calf intestinal alkaline phosphatase, an acid phosphatase, or a recombinant version of such a phosphatase in combination with a chromogenic substrate such as 5-bromo-4-chloro-3-indolyl phosphate (BCIP), 6-chloro-3-indolyl phosphate, 5-bromo-6-chloro-3-indolyl phosphate, p-nitrophenyl phosphate, or o-nitrophenyl phosphate or with a fluorogenic substrate such as 4-methylumbelliferyl phosphate, carboxyumbelliferyl phosphate, 6,8-difluoro-7-hydroxy4-methylcoumarinyl phosphate (DiFMUP, U.S.
  • a fluorogenic substrate such as 4-methylumbelliferyl phosphate, carboxyumbelliferyl phosphate, 6,8
  • Glycosidases in particularly ⁇ -galactosidase, ⁇ -glucuronidase, and ⁇ -glucosidase, are additional suitable enzymes.
  • Appropriate chromogenic substrates include, but are not limited to, 5-bromo-4-chloro-3-indolyl ⁇ -D-galactopyranoside (X-gal) and similar indolyl galactosides, glucosides, and glucuronides, o-nitrophenyl ⁇ -D-galactopyranoside (ONPG), and p-nitrophenyl ⁇ -D-galactopyranoside.
  • Preferred fluorogenic glycosidase substrates include resorufm ⁇ -D-galactopyranoside, fluorescein digalactoside (FDG), fluorescein diglucuronide and their structural variants (U.S. Pat. Nos. 5,208,148; 5,242,805; 5,362,628; 5,576,424 and 5,773,236), 4-methylumbelliferyl ⁇ -D-galactopyranoside, carboxyumbelliferyl ⁇ -D-galactopyranoside, and fluorinated coumarin ⁇ -D-galactopyranosides (U.S. Pat. No. 5,830,912).
  • Additional enzymes include, but are not limited to, hydrolases such as cholinesterases and peptidases, oxidases such as glucose oxidase and cytochrome oxidases, and reductases, for which suitable substrates are known.
  • Enzymes and their appropriate substrates that produce chemiluminescence are preferred for some assays. These include, but are not limited to, natural and recombinant forms of luciferases and aequorins and include their chimeras with proteins that include protein A, protein G, and protein L. Chemiluminescence-producing substrates for phosphatases, glycosidases, and oxidases such as those containing stable dioxetanes, luminol, isoluminol, and acridinium esters are additionally useful.
  • the sample is illuminated at a suitable absorption wavelength.
  • a suitable wavelength is one that comes within the range of absorption wavelengths for each of the fluorescent dyes being used.
  • the mixture is illuminated by a light source capable of producing light at or near the wavelength of maximum absorption of the dye or dyes, such as by ultraviolet or visible lamp, an arc lamp, a laser, or even sunlight. Illumination of the sample at a suitable wavelength results in one or more illuminated targets that are then analyzed according to the response of their fluorescence to the illumination.
  • the illuminated targets are observed with any of a number of means for detecting a fluorescent response emitted from the illuminated target, including but not limited to visual inspection, cameras and film or other imaging equipment, or use of instrumentation such as fluorometers, plate readers, laser-based scanners, microscopes, or flow cytometers, or by means for amplifying the signal such as a photomultiplier (PMT).
  • means for detecting a fluorescent response emitted from the illuminated target including but not limited to visual inspection, cameras and film or other imaging equipment, or use of instrumentation such as fluorometers, plate readers, laser-based scanners, microscopes, or flow cytometers, or by means for amplifying the signal such as a photomultiplier (PMT).
  • PMT photomultiplier
  • the labeling mixture is combined with a sample of cells in a fluid, such as ascites, hybridoma supernatant, or serum, and the presence or absence of the target in such cells is detected by using an automated instrument that sorts cells according to the detectable fluorescence response of the detectable moieties in the immunolabeling complexes bound to such cells, such as by fluorescence activated cell sorting (FACS), which is described in Mansour, et al., U.S. Pat. No. 4,665,024 (1987) and is incorporated by reference.
  • FACS fluorescence activated cell sorting
  • additional detection reagents are combined with the sample concurrently with or following the labeling mixture.
  • Such additional detection reagents include, but are not limited to reagents that selectively detect cells or subcellular components, ions, or indicate the cell viability, life cycle, or proliferation state.
  • the additional detection reagent is a labeled antibody that is directly or indirectly detectable and another additional detection reagent is a stain for nucleic acids, for F-actin, or for a cellular organelle.
  • Determination of more than one target in a sample is accomplished with a labeling mixture containing more than one immunolabeling complex wherein a first target-binding antibody is bound to a labeling proteins with a first label and a second target-binding protein is bound to a labeling protein with a second label, wherein the first and second labels are detectably distinct.
  • a labeling mixture containing more than one immunolabeling complex
  • a first target-binding antibody is bound to a labeling proteins with a first label
  • a second target-binding protein is bound to a labeling protein with a second label
  • the first and second labels are detectably distinct.
  • compositions of the present invention include isolated immunolabeling complexes and isolated monovalent labeling proteins.
  • the labeling protein is labeled with one or more labels.
  • the labels that are attached to the labeling protein are directly detectable moieties, which directly detectable moieties are optionally the same or different, or the label is an enzyme or a hapten that is indirectly detectable.
  • the labeling protein is prepared by the process of (a) complexation of the unlabeled labeling protein with an immobilized form of its complementary binding protein; and (b) chemical conjugation of the labeling protein to the label; and (c) disaggregation of the covalently labeled labeling protein from the immobilized complementary protein.
  • Covalent conjugates of the enzyme and the labeling protein of the invention that are suitable for preparation of labeling complexes are typically prepared by methods well known in the art (MOLECULAR PROBES HANDBOOK OF FLUORESCENT PROBES AND RESEARCH CHEMICALS, Chapter 5 (1996)). Additionally, fusion proteins of enzymes such as of alkaline phosphatase, horseradish peroxidase, luciferase, and aequorin with the labeling proteins, protein A and protein G, have been described (Sun et al., J. Immunol. Meth. 152, 43 (1992); Eliasson et al., J. Biol. Chem.
  • Chemical labeling of the monovalent antibody fragment is optionally performed while the antibody fragment is immobilized on a matrix, such as while bound to its complementary binding site.
  • a matrix such as while bound to its complementary binding site.
  • an Fab fragment of goat anti-(mouse Fc) or a protein A or protein G can be bound to a whole mouse IgG that has been immobilized on agarose.
  • a reactive dye or coupling to a protein such as a phycobiliprotein, a tandem conjugate of a phycobiliprotein with an Alexa Fluor dye (Molecular Probes, Inc.) or with an enzyme by standard methods, the excess reactive dye is removed while the monovalent labeling protein is still immobilized and then the labeled monovalent antibody is eluted by reducing the pH.
  • This method has the simultaneous advantages of facilitating elimination of the unconjugated reactive dye and protecting the combining region of the monovalent antibody. Furthermore, we have observed that fluorescent dye conjugates prepared by this indirect method typically have greater fluorescence or activity than do conjugates of the monovalent labeling protein prepared using the same reactive dye in a homogeneous solution. We have also observed that less labeled monovalent labeling protein is required to achieve the same or superior detectability when preparing a labeling complex with the target-binding antibody. Alternatively, smaller quantities of target-binding antibody can be used when pre-forming the immunolabeling complexes to achieve the same detectablity. Optionally, the target-binding antibody is about 5 micrograms to about 0.001 micrograms.
  • the invention also includes an isolated protein G complex consisting essentially of protein G and a non-covalently bound albumin.
  • the albumin is covalently labeled with one or more detectable labels, as described above, where the detectable labels are optionally the same or different.
  • detectable labels are optionally the same or different.
  • Various types of albumin can be used including, for example, human albumin, bovine serum albumin, or ovalbumin.
  • the isolated immunolabeling complex is made by combining, in vitro, one or more target-binding antibodies with an excess of labeling protein that has been covalently labeled as described above, under conditions suitable for the labeling protein to bind selectively and with high affinity to a selected region of said target-binding antibody to form an immunolabeling complex, and isolating said immunolabeling complex.
  • combining means mixing a solution of the isolated covalently labeled labeling protein with a solution of the target-binding antibody in a suitable buffer for a period sufficient to form a noncovalent complex. Typically this period is under five minutes but may be less than one minute.
  • the labeling protein is mixed in a molar ratio of at least one to 50 moles of labeling protein to one mole of the region of the target-binding antibody to be complexed. More commonly a ratio of at least one to as many as 10 moles of labeling protein per mole of target-binding antibody is combined.
  • a molar ratio of approximately 2 to 10 is typical, more typically 3 to 5 (particularly for complexes in which the labeling protein has been labeled while immobilized on an affinity matrix).
  • the ease of formation of the complex permits rapid optimization of the complex and assessment of the effect of variation in experimental parameters.
  • a particularly unique advantage of the invention is that the stoichiometry of the complex is easily adjusted to provide complexes with different ratios of labeling protein to target-binding antibody, and thus there is some control over the ultimate detectability of the target in the sample. This feature is impractical with primary antibodies that have been directly chemically labeled with dyes. Complexes that have been labeled with the same dye but at different molar ratios can be separately detected by the differences in their intensities.
  • a unique and important advantage of the method of the invention is the ability to form the immunolabeling complex from exceedingly small quantities of the target-binding antibody and labeling protein, which is impractical when using chemically reactive dyes.
  • Complexes of the invention have been prepared from sub-micrograms of a target-binding antibody with sub-micrograms of a labeling protein. The process of forming a labeling complex can easily be automated. An important use of this characteristic is that of screening hybridoma supernatants in the generation and optimization of new antibodies, where the very small quantity of antibody present in a single well makes it impractical to perform chemical labeling.
  • a capture component is added to the labeling mixture, prior to combining the labeling mixture with the sample, to remove excess labeling protein.
  • a capture component For applications in which immunolabeling complexes of multiple primary antibodies from the same species (e.g. mouse monoclonal antibodies) or cross-reacting species (e.g. mouse and human antibodies) are to be used simultaneously or sequentially, it is necessary to quench or otherwise remove any excess labeling protein by use of a capture component or by other means to avoid inappropriate labeling of the sample.
  • the most effective capturing components to capture excess labeling protein are those that contain the binding site of the labeling protein but are themselves not labeled, preferably an antibody or antibody fragment.
  • Capture components may be free in solution or immobilized on a solid phase, such as agarose, cellulose, or a natural or synthetic polymer, to facilitate separation of the excess capture component from the labeled protein.
  • the capture component is optionally attached to a microsphere or magnetic particle.
  • Any of the isolated labeling mixtures optionally include a capture component to remove excess labeling protein.
  • separation of excess labeling protein is not essential for successful utilization of the invention, particularly when using a single target-binding antibody.
  • labeling mixtures comprising one or more immunolabeling complexes, wherein at least one immunolabeling complex is comprised of a target-binding antibody, is selective for a target in a sample, and a labeling protein, as described above, that binds selectively and with high affinity to a selected region of the target-binding antibody. More preferably the labeling mixture contains at least three such complexes.
  • the potential number of immunolabeling complexes that can be used simultaneously is principally limited by the number of targets for which selective antibodies are available and the ability to spatially or spectrally resolve the detectable signals, such as on a Western blot, a protein array, a chromosome “spread”, or a microsphere or “chip” array (e.g., U.S. Pat. Nos. 5,981,180 and 5,736,330).
  • labeling mixtures optionally include additional detection reagents such as those known to those skilled in the art, including reagents that localize the same or additional targets in the sample.
  • additional detection reagents include stains for cell organelles, reagents to indicate the state of cell viability or proliferation, and enzyme substrates.
  • Other preferred additional detection reagents are additional antibodies that can be detected in conjunction with direct or indirect labeling methods, wherein the targets of such additional antibodies and of the target-binding antibody are optionally the same or different.
  • the labeling protein is a monovalent fragment that is made by (a) generating a labeling antibody against an antigen, where said antigen is a Fc region of one or more isotypes of an antibody from a different species; (b) digesting said labeling antibody to generate multiple monovalent fragments; (c) covalently labeling said monovalent fragments with one or more labels; and (d) isolating the labeled fragments.
  • mouse or rat monoclonal antibodies is an Fab fragment of an antibody to mouse Fc, wherein the whole antibody was produced in goat, chicken or other species.
  • Preferred as a labeling protein for complexing with either targeting or secondary antibodies produced in other species are antibodies to the Fc region of these target-binding or secondary antibodies produced in a non-cross-reacting animal, in particular Fab fragments of such antibodies.
  • additional selectivity can be obtained by utilizing an antibody directed at a particular isotype of the primary antibody, such as toward IgG 1 , IgG 2a , or IgG 3 isotypes.
  • the target-binding antibody is selected based on the desired target, where the target is an antigen selected for investigation or analysis.
  • the chemical identity of the target antigen may be known or unknown.
  • the target is optionally a material of biological or synthetic origin that is present as a molecule or as a group of molecules. Typically, the target is a biological material or antigenic determinant.
  • Bio materials include, but are not limited to, antibodies, amino acids, proteins, peptides, polypeptides, enzymes, enzyme substrates, hormones, lymphokines, metabolites, antigens, haptens, lectins, avidin, streptavidin, toxins, poisons, environmental pollutants, carbohydrates, oligosaccharides, polysaccharides, glycoproteins, glycolipids, nucleotides, oligonucleotides, nucleic acids and derivatized nucleic acids (including deoxyribo- and ribonucleic acids and peptide nucleic acids), DNA and RNA fragments and derivatized fragments (including single and multi-stranded fragments), natural and synthetic drugs, receptors, virus particles, bacterial particles, virus components, biological cells, cellular components (including cellular membranes and organelles), natural and synthetic lipid vesicles, and polymer membranes.
  • the target material is present as a component or contaminant of a sample taken from a biological or environmental
  • kits for preparing a labeling protein, for preparing an immunolabeling complex, and for immunolabeling a target in a sample also form part of the invention.
  • kits can be prepared from readily available materials and reagents and can come in a variety of embodiments. The contents of the kit will depend on the design of the assay protocol or reagent for detection or measurement. Generally, the kits will contain instructions, appropriate reagents and labels, and solid supports, as needed.
  • instructions include a tangible expression describing the reagent concentration or at least one assay method parameter such as the relative amounts of reagent and sample to be admixed, maintenance time periods for reagent/sample admixtures, temperature, buffer conditions and the like to allow the user to carry out any one of the methods or preparations described above.
  • a kit is designed for a simple detection assay for a target using a single highly specific antibody
  • the kit contains this highly specific antibody pre-complexed with a labeling protein.
  • the kit comprises panels of antibodies each pre-complexed with a unique labeling protein whose recognition pattern for the desired target is calibrated or the kit comprises panels of antibodies and a separate labeling protein or labeling proteins.
  • a labeled competitive mimotope or mixture is optionally additionally included to permit the assays to be conducted by competition of the unlabeled target in the sample with the labeled mimotope.
  • a kit for preparation of optimal labeling proteins typically comprises (a) a monovalent antibody fragment that binds to a specific region of a target-binding antibody, (b) a chemically reactive dye, and (c) an immobilization matrix comprising the specific region of the target-binding antibody to which the monovalent antibody fragment binds.
  • the kit comprises (a) a mixture of one or more monovalent antibody fragments that bind to a specific region of one or more antibody isotypes, where each fragment is covalently labeled with one or more labels, (b) a capture component to which the antibody fragments bind, and optionally (c) comprises one or more target-binding antibodies.
  • the capture component is purified mouse IgG or serum from a non-immune mouse and the label is a fluorescent molecule.
  • the antibody fragment is an antibody to the Fc region of the target-binding antibody that is matched to or cross-reacts with the species from which the target-binding antibody is derived.
  • the labeled monovalent antibody fragment has been labeled while immobilized on an affinity matrix.
  • a kit for preparing an immunolabeling complex comprising another version of the kit for preparing (a) an immunolabeling complex that comprises a protein G complexed with a labeled albumin and (b) a capture component to which the protein G complexed with albumin binds.
  • the capture component is purified mouse IgG or non-immune mouse serum and the albumin is human albumin, bovine serum albumin, or ovalbumin. In a more preferred embodiment the albumin is ovalbumin.
  • the kit contains three or more such immunolabeling complexes.
  • kits and components can be prepared according to the present invention, depending upon the intended user of the kit and the particular needs of the user.
  • the instant invention has useful applications in basic research, high-throughput screening, immunohistochemistry, fluorescence in situ hybridization (FISH), microarray technology, diagnostics, and medical therapeutics.
  • the invention can be used in a variety of assay formats for diagnostic applications in the disciplines of microbiology, immunology, hematology and blood transfusion, tissue pathology, forensic pathology, and veterinary pathology.
  • the invention is particularly useful in the characterization and selection of optimized antibodies from hybridoma supernatants.
  • the invention can be used to deliver therapeutics to a specific target.
  • the current invention provides a versatile and convenient method to enhance any assay that uses an antibody as part of its detection methodology.
  • the instant invention can be used to study biological phenomena, such as, for example, cell proliferation, signal transduction in cells, or apoptosis.
  • biological phenomena such as, for example, cell proliferation, signal transduction in cells, or apoptosis.
  • BrdU is a marker for both cell proliferation and apoptosis, as it is readily incorporated into newly synthesized DNA that has progressed through the S-phase of the cell cycle and also into DNA break sites by deoxynucleotidyl transferase (TdT).
  • Anti-BrdU antibodies are used to detect cells marked by BrdU incorporation. By being able to directly label the anti-BrdU antibodies, the current invention provides a convenient method to allow for detection of the incorporated BrdU by conventional immunohistochemistry or fluorescence, depending on detection method required.
  • the current invention has the advantage of allowing staining for multiple targets in one cocktail, thereby reducing the need for more samples or processing steps per experiment. This is particularly important when analyzing precious samples (e.g., pediatric samples, leukocytes isolated from biopsies, rare antigen-specific lymphocytes and mouse tissues that yield a small number of cells).
  • precious samples e.g., pediatric samples, leukocytes isolated from biopsies, rare antigen-specific lymphocytes and mouse tissues that yield a small number of cells.
  • labeled secondary antibodies directed at individual isotype-specific targeting antibodies e.g., anti-IgG 1 isotype antibodies
  • this type of labeled antibody it is not possible to use this type of labeled antibody to detect more than one of the same isotype of an antibody (e.g., an IgG 1 isotype antibody) in a single sample due to cross-reactivity.
  • the current invention overcomes these limitations by providing for a convenient and extremely versatile method of rapidly labeling either small or large quantities of any primary antibody including primary antibodies of the same isotype to be used in, for example, multicolor flow cytometry and on Western blots. This advance in multicolor systems has a number of advantages over current two- and three-color flow cytometric measurements.
  • no combination of one-color stains can accurately enumerate or be used to isolate CD3 + CD4 + CD8 ⁇ T cells (excluding, for example CD3 + CD4 + CD8 + T cells and small CD4 + monocytes).
  • CD3 + CD4 + CD8 + T cells excluding, for example CD3 + CD4 + CD8 + T cells and small CD4 + monocytes.
  • the use of cell membrane markers to study leukocyte composition in blood and tissue serves as an example of an analytical monoclonal antibody application, particularly in combination with flow cytometry. It is also the example most relevant to studies of the immune system, because the cellular composition of blood and lymphoid tissue provides a ‘window’, allowing the analysis and monitoring of the immune system.
  • the methods of the invention can also be used in immunofluorescence histochemistry.
  • Immunofluorescence histochemistry involves the use of antibodies labeled with fluorophores to detect substances within a specimen.
  • the pathologist derives a great deal of information of diagnostic value by examining thin sections of tissue in the microscope. Tissue pathology is particularly relevant to, for example, the early diagnosis of cancer or premalignant states, and to the assessment of immunologically mediated disorders, including inflammation and transplant rejection.
  • the problems associated with immunofluorescence histochemistry stem from the limitations of the methods currently available for use in such application. For example, directly labeling an antibody can result in antibody inactivation and requires a relatively large of amount of antibody and time to do the conjugation.
  • BrdU-labeled DNA in rodent tissue is detected by immunohistochemical staining.
  • the target-binding antibody is conventionally mouse anti-BrdU, and the detecting antibody system uses an anti-mouse immunoglobulin antibody, labeled with fluorescein. Because there is homology between mouse immunoglobulin and immunoglobulins from a number of rodent species (for example, rats, mice, hamsters, etc.), the detecting antibody not only binds to the target-binding antibody, but also nonspecifically binds to immunoglobulin in the tissue.
  • the current invention eliminates this problem by pre-forming the immunolabeling complex and allows for a simple, rapid and convenient method to proceed with labeling with two, three or more fluorescent antibodies in one experiment. Very significantly, it can always be used with primary antibodies of either the same or different isotype, and always on tissue of the same or similar species as the primary antibody.
  • the instant invention also has application in the field of microarrays.
  • Microarray technology is a powerful platform for biological exploration (Schena (Ed.), Microarray Biochip Technology, (2000)).
  • Many current applications of arrays, also known as “biochips,” can be used in functional genomics as scientists seek characteristic patterns of gene expression in different physiopathological states or tissues.
  • a common method used in gene and protein microarray technology involves the use of biotin, digoxigenin (DIG), or dinitrophenyl (DNP) as an epitope or a “tag” such as an oligohistidine, glutathione transferase, hemagglutinin (HA), or c-myc.
  • a detectably labeled anti-biotin, anti-DIG, anti-DNP, anti-oligohistidine, anti-glutathione transferase, anti-HA, or anti-c-myc is used as the detection reagent.
  • the instant invention allows for the use of multiple fluorophore- or enzyme-labeled antibodies, thereby greatly expanding the detection modalities and also providing for enhanced multiplexing and two-dimensional analysis capabilities.
  • the invention can be used with protein microarrays and on Western blots.
  • Protein microarrays can provide a practical means to characterize patterns of variation in hundreds of thousands of different proteins in clinical or research applications.
  • Antibody arrays have been successfully employed that used a set of 115 antibody/antigen pairs for detection and quantitation of multiple proteins in complex mixtures (Haab et al., Genome Biology, 2, 4.1 (2001)).
  • protein microarrays use very low sample volumes, which historically have significantly limited the use of antibody technology for this application.
  • the invention of the application readily overcomes this limitation and provides a means to label antibodies with the fluorescent dyes using a very low sample volume and to automate formation of the staining complex and the staining process.
  • the present invention also provides a means for the specific detection, monitoring, and/or treatment of disease and contemplates the use of immunolabeling complexes to detect the presence of particular targets in vitro.
  • the sample may be utilized in liquid phase, in a gel, or bound to a solid-phase carrier, such as an array of fluorophore-labeled microspheres (e.g., U.S. Pat. No. 5,981,180 and 5,736,330).
  • a sample can be attached to a polymer, such as aminodextran, in order to link the sample to an insoluble support such as a polymer-coated bead, plate, or tube.
  • a polymer such as aminodextran
  • the immunolabeling complexes of the present invention can be used to detect the presence of a particular target in tissue sections prepared from a histological specimen.
  • the tissue to be assayed will be obtained by surgical procedures, e.g., biopsy.
  • the excised tissue will be assayed by procedures generally known in the art, e.g. immunohistochemistry, for the presence of a desired target that is recognized by an immunolabeling complex, as described above.
  • the tissue may be fixed or frozen to permit histological sectioning.
  • the immunolabeling complex may be labeled, for example with a dye or fluorescent label, chemical, heavy metal or radioactive marker to permit the detection and localization of the target-binding antibody in the assayed tissue.
  • In situ detection can be accomplished by applying a detectable immunolabeling complex to the tissue sections. In situ detection can be used to determine the presence of a particular target and to determine the distribution of the target in the examined tissue: General techniques of in situ detection are well known to those of ordinary skill. See, for example, Ponder, “Cell Marking Techniques and Their Application,” in MAMMALIAN DEVELOPMENT: A PRACTICAL APPROACH, Monk (ed.), 115 (1987).
  • tissues are probed with an immunolabeling complex, as defined above, that comprises a target-binding antibody to a target antigen associated with the disease, e.g., by immunohistochemical methods.
  • an immunolabeling complex comprising a target-binding antibody to the disease antigen are preferably used in immunoassays to detect a secreted disease antigen target.
  • Detection can be by a variety of methods including, for example, but not limited to, flow cytometry and diagnostic imaging.
  • flow cytometry for the detection method, the use of microspheres, beads, or other particles as solid supports for antigen-antibody reactions in order to detect antigens or antibodies in serum and other body fluids is particularly attractive.
  • Flow cytometers have the capacity to detect particle size and light scattering differences and are highly sensitive fluorescence detectors. Microfluidic devices provide a means to perform flow-based analyses on very small samples.
  • diagnostic imaging can be used.
  • the method of diagnostic imaging with radiolabeled antibodies is well known. See, for example, Srivastava (ed.), RADIOLABELED MONOCLONAL ANTIBODIES FOR IMAGING AND THERAPY, Plenum Press (1988); Chase, “Medical Applications of Radioisotopes,” in REMINGTON'S PHARMACEUTICAL SCIENCES, 18th Edition, Gennaro et al. (eds.) Mack Publishing Co., 624 (1990); and Brown, “Clinical Use of Monoclonal Antibodies,” in BIOTECHNOLOGY AND PHARMACY, Pezzuto et al. (eds.), Chapman & Hall, 227 (1993).
  • This technique also known as immunoscintigraphy, uses a gamma camera to detect the location of gamma-emitting radioisotopes conjugated to antibodies. Diagnostic imaging is used, in particular, to diagnose cardiovascular disease and infectious disease.
  • the present invention contemplates the use of immunolabeling complexes to diagnose cardiovascular disease.
  • immunolabeling complexes comprising anti-myosin antibodies can be used for imaging myocardial necrosis associated with acute myocardial infarction.
  • Immunolabeling complexes comprising antibodies that bind platelets and fibrin can be used for imaging deep-vein thrombosis.
  • immunolabeling complexes comprising antibodies that bind to activated platelets can be used for imaging atherosclerotic plaque and immunolabeling complexes.
  • Immunolabeling complexes of the present invention also can be used in the diagnosis of infectious diseases.
  • immunolabeling complexes comprising antibodies that bind specific bacterial antigens can be used to localize abscesses.
  • immunolabeling complexes comprising antibodies that bind granulocytes and inflammatory leukocytes can be used to localize sites of bacterial infection.
  • the immunolabeling complexes of the present invention can be used to detect signal transduction in cells, the products of signal transduction, and defects, inhibitors, and activators of signal transduction.
  • the present invention contemplates the detection of cancer using immunolabeling complexes comprising antibodies that bind tumor markers (targets) to detect cancer.
  • tumor markers include carcinoembryonic antigen, ⁇ -fetoprotein, oncogene products, tumor-associated cell surface antigens, and necrosis-associated intracellular antigens.
  • antibody imaging can be used to monitor therapeutic responses, detect recurrences of a disease, and guide subsequent clinical decisions and surgical procedures.
  • fluorescent complexes that absorb and emit light in the near infrared (such as those of the Alexa Fluor 700 and Alexa Fluor 750 dyes) is also known.
  • Immunolabeling complexes that are therapeutically substituted or therapeutic immunolabeling complexes, wherein the labeling protein is alternatively conjugated with a radioactive moiety, a toxin, or a drug, as opposed to with a detectable label, to form a drug-labeled protein or immunoconjugate, can be used to treat viral and bacterial infectious diseases, cardiovascular disease, autoimmune disease, and cancer.
  • the objective of such therapy is to deliver cytotoxic doses of radioactivity, toxin, or drug to target cells, while minimizing exposure to non-target tissues.
  • Therapeutic agents useful in the treatment of a disease may be conjugated to the protein by methods known to those of skill in the art.
  • therapeutic agents include peptides; proteins; small organic drugs, such as, doxorubicin, daunorubicin, methotrexate, melphalin, chlorambucil, vinca alkaloids, 5-fluorouridine, and mitomycin-C; radioisotopes, such as, 131 I, 67 Cu, or 90 Y; and cytotoxic agents, such as ricin, abrin, pokeweed antiviral protein, gelonin, diphtherin toxin, and Pseudomonas endotoxin.
  • 131 I can be covalently bound to the protein by methods known in the art, e.g., lodogen reactions (Frank et al., Biochem. Biophys. Res. Commun. 80, 849 (1978)).
  • 67 Cu one of the more promising radioisotopes for radioimmunotherapy due to its 61.5 hour half-life and abundant supply of beta particles and gamma rays, can be conjugated to the protein using the chelating agent, 6-(p-bromoacetamidobenzyl)-1,4,8,1 1-tetraazacyclotetradecane-N,N′,N′′,N′′′-tetraacetic acid (TETA).
  • TETA 6-(p-bromoacetamidobenzyl)-1,4,8,1 1-tetraazacyclotetradecane-N,N′,N′′,N′′′-tetraacetic acid
  • 90 Y which emits an energetic beta particle, can be coupled to a labeling protein using diethylenetriaminepentaacetic acid (DTPA), or more preferably, 1,4,7,10-tetraazacyclododecane-N,N′,N′′,N′′′-tetraacetic acid (DOTA) (Camera et al., J. Nucl. Med. 35, 882 (1994)).
  • DTPA diethylenetriaminepentaacetic acid
  • DOTA 1,4,7,10-tetraazacyclododecane-N,N′,N′′,N′′′-tetraacetic acid
  • Agents such as glutaraldehyde or a carbodimide may conjugate the toxins to the protein.
  • Small organic drugs, peptides, and proteins can be conjugated by various means known in the art, for example, but not limitation, through amide bonds and or linkers.
  • Administration of the therapeutically substituted immunolabeling complex may be by various means known in the art, but generally will be by injection, intravenous, intraarterial, intraperitoneal, intramuscular, subcutaneous, intrapleural, intrathecal, by perfusion through a regional catheter, or by direct intralesional injection.
  • the administration may be by continuous infusion, or by single or multiple boluses.
  • the therapeutic immunolabeling complex of the present invention can be formulated according to known methods to prepare pharmaceutically useful compositions, whereby the therapeutic immunolabeling complexes are combined in a mixture with a pharmaceutically acceptable carrier.
  • a composition is said to be a “pharmaceutically acceptable carrier” if its administration can be tolerated by a recipient patient.
  • Sterile phosphate-buffered saline is one example of a pharmaceutically acceptable carrier.
  • Other suitable carriers are well known to those in the art. See, for example, REMINGTON'S PHARMACEUTICAL SCIENCES, 18th Ed. (1990).
  • a substituted immunolabeling complex and a pharmaceutically acceptable carrier are administered to a patient in a therapeutically effective amount.
  • a combination of an immunolabeling complex and a pharmaceutically acceptable carrier is said to be administered in a “therapeutically effective amount” if the amount administered is physiologically significant.
  • An agent is physiologically significant if its presence results in a detectable change in the physiology of a recipient patient.
  • a useful therapeutic dose will vary with the particular therapeutic agent used, the particular disease type and history, and the specific considerations of the patient to be treated, such as the patient's age, weight, height, sex, general medical condition, and previous medical history.
  • a physician administering the agent will know to calculate the effective therapeutic dose, which dose will be effective in reducing or eliminating the disease without compromising significantly normal tissues or cells of the patient.
  • a dosage of the substituted immunolabeling complex that is in the range of from about 1 pg/kg to 10 mg/kg (amount of agent/body weight of patient), although a lower or higher dosage may also be administered.
  • many studies have demonstrated successful treatment with doses of 0.1 to 1.0 milligram, while other studies have shown improved localization with doses in excess of 10 milligrams.
  • Controlled release preparations can be prepared through the use of polymers to complex or adsorb a substituted immunolabeling complex.
  • biocompatible polymers include matrices of poly(ethylene-co-vinyl acetate) and matrices of a polyanhydride copolymer of a stearic acid dimer and sebacic acid (Sherwood et al., Biotechnology 10, 1446 (1992).
  • the rate of release of a substituted immunolabeling complex from such a matrix depends upon the molecular weight of the substituted immunolabeling complex, the amount of substituted immunolabeling complex within the matrix, and the size of dispersed particles (Saltzman et al., Biophysical J. 55, 163 (1989). Other solid dosage forms are described in REMINGTON'S PHARMACEUTICAL SCIENCES, 18th Ed. (1990). Each of the above-described references is incorporated herein by reference in its entirety.
  • Purified mouse IgG was fragmented with the proteolytic enzyme papain (CURRENT PROTOCOLS IN CELL BIOLOGY, 16.4.1-16.4.10 (2000)).
  • a 12 mL solution of mouse IgG was prepared at ⁇ 2 mg/mL in phosphate-buffered saline (PBS).
  • a solution containing 0.1 mg of papain in digestion buffer (PBS, 0.02 M EDTA, 0.02 M cysteine) was added to the antibody and allowed to react at 37° C. for 16 hours. The digestion was terminated by the addition 20 ⁇ L of 0.3 M iodoacetamide in PBS. The fragments were dialyzed against 2 L of PBS for 16 hours at 4° C.
  • the Fc fragment was purified on a protein G-Sepharose CL-4B column.
  • the bound fraction containing the Fc fragment was eluted from the column using 50-100 mM glycine/HCl buffer, pH 2.5-2.8.
  • the eluate was collected in 1 mL fractions.
  • the pH of the protein fractions was immediately raised to neutral by addition of 100 ⁇ L of either 500 mM phosphate or Tris buffer, pH 7.6, to each 1 mL fraction.
  • the solution was then loaded onto a Sephacryl S-200 Superfine size-exclusion column and fractions corresponding to a molecular weight of ⁇ 50 kDa were collected and analyzed by SDS-PAGE and HPLC.
  • the goat anti-(rabbit Fc) fragment was prepared similarly.
  • the immunogen used to immunize the animal was the purified mouse or rabbit IgG Fc domain (see Example 1).
  • Methods of immunizing animals are well known and conventional, and suitable immunization protocols and immunogen concentrations can be readily determined by those skilled in the art (Current Protocols in Immunology 2.4.1-9 (1995); ILAR Journal 37, 93 (1995)). Briefly, individual goats were immunized with purified mouse Fc or purified rabbit Fc. The initial immunization in 50% Freund's complete adjuvant (1000 ⁇ g conjugate (half subcutaneous, half intramuscularly)) was followed by 500 ⁇ g conjugate per goat in Freund's incomplete adjuvant two and four weeks later and at monthly intervals thereafter.
  • Antibodies were harvested from serum using protein A-Sepharose chromatography. Antibodies against mouse Fc isotypes can be prepared by starting with isotype-selected mouse Fc antigens. Rabbits have a single Fc isotype. Characterization of the selectivity and cross-reactivity of isotype-specific antibodies is by standard techniques, including HPLC.
  • Conjugates of antibody immunoglobulin-binding protein or the non-antibody immunoglobulin-binding peptides or proteins with low molecular weight dyes and haptens such as biotin or digoxigenin are typically prepared from succinimidyl esters of the dye or hapten, although reactive dyes and haptens having other protein-reactive functional groups are also suitable.
  • the typical method for protein conjugation with succinimidyl esters is as follows. Variations in molar ratios of dye-to-protein, protein concentration, time, temperature, buffer composition and other variables that are well known in the art are possible that still yield useful conjugates.
  • a protein solution of the Fab fragment of goat anti-(rabbit Fc), goat anti-(mouse Fc), protein A, protein G, or protein L or an immunoglobulin-binding peptide (e.g., a peptide identified by screening a library of peptides) is prepared at ⁇ 10 mg/mL in 0.1 M sodium bicarbonate (pH ⁇ 8.3).
  • the labeling reagents are dissolved in a suitable solvent such as DMF at ⁇ 10 mg/mL. Predetermined amounts of the labeling reagents are added to the protein solution with stirring.
  • a molar ratio of 10 moles of dye to 1 mole of protein is typical, though the optimal amount can be varied with the particular labeling reagent, the protein being labeled and the protein's concentration. The optimal ratio was determined empirically. When optimizing the fluorescence yield and determining the effect of degree of substitution (DOS) on the conjugate's brightness, it is typical to vary the ratio of reactive dye to protein over a several-fold range.
  • the reaction mixture is incubated at room temperature for a period that is typically one hour or on ice for several hours.
  • the dye-protein conjugate is typically separated from unreacted reagents by size-exclusion chromatography, such as on BIO-RAD P-30 resin equilibrated with PBS.
  • the initial, protein-containing band is collected and the DOS is determined from the absorbance at the absorbance maximum of each fluorophore, using the extinction coefficient of the free fluorophore.
  • the DOS of nonchromophoric labels, such as biotin is determined as described in Haugland (Haugland et al., Meth. Mol. Biol. 45, 205 (1995); Haugland, Meth. Mol. Biol. 45, 223 (1995); Haugland, Meth. Mol. Biol. 45, 235 (1995); Haugland, Current Protocols in Cell Biol. 16.5.1-16.5.22 (2000)).
  • conjugates of goat anti-(mouse Fc) and goat anti-(rabbit Fc) were prepared with several different Alexa Fluor dyes, with Oregon Green dyes, with biotin-X succinimidyl ester, with desthiobiotin-X succinimidyl ester, with succinimidyl 3-(2-pyridyldithio)propionate (SPDP) and with succinimidyl trans-4-(maleimidylmethyl)cyclohexane-1-carboxylate (SMCC).
  • Some dye conjugates of protein A and protein G are commercially available, such as from Molecular Probes. Inc. (Eugene, Oreg.).
  • the interspecies specificity and approximate affinity of some other non-antibody immunoglobulin-binding proteins bind to segments of a target antibody, such as that of protein A and protein G are known (Langone, Adv. Immunol. 32, 157 (1982); Surolia et al., Trends Biochem. Sci. 7, 74 (1982); Notani et al., J. Histochem. Cytochem. 27, 1438 (1979); Goding, J. Immunol. Meth. 20, 241 (1978); J. Immunol. Meth. 127, 215 (1990); Bjorck et al., J. Immunol. 133, 969 (1984)).
  • labeling proteins (goat Fab anti-(mouse Fc), goat Fab anti-(mouse lambda light chain), goat Fab anti-(mouse kappa light chain), protein A, protein G, protein L, lectins, single-chain fragment variable antibodies (ScFv) ) conjugated to the detectable labels of R-phycoerythrin (R-PE), allophycocyanin (APC), tandem conjugates of phycobiliproteins with chemical dyes including several Alexa Fluor dyes, horseradish peroxidase (HRP), Coprinus cinereus peroxidase, Arthromyces ramosus peroxidase, glucose oxidase and alkaline phosphatase (AP) were or can be prepared by standard means (Haugland et al., Meth.
  • Fusion proteins such as of protein G or protein A with detectable labels such as luciferin, aequorin, green-fluorescent protein and alkaline phosphatase are also known that are suitable for practice of the invention (Sun et al., J. Immunol. Meth. 152, 43 (1992); Eliasson et al., J. Biol. Chem. 263, 4323 (1988); Eliasson et al., J. Immunol. 142, 575 (1989)).
  • Immunoglobulin heavy and light chains like most secreted and membrane bound proteins, are synthesized on membrane-bound ribosomes in the rough endoplasmic endoplasmic reticulum where N-linked glycosylation occurs.
  • the specificity of lectins for carbohydrates, including N-linked glycoproteins, is also known (EY laboratories, Inc. Lectin Conjugates Catalog, 1998).
  • Unlabeled Fab fragment for goat anti-(mouse Fc) (prepared as in Example 3) was bound to agarose-immobilized mouse IgG for one hour. Following a wash step with bicarbonate buffer, pH 8.3, the complex of immobilized IgG and unlabeled Fab was labeled for one hour at room temperature with the succinimidyl ester of the amine-reactive label. Unconjugated dye was eluted with bicarbonate buffer, and then the covalently labeled Fab fragment was eluted with 50-100 mM glycine/HCl buffer, pH 2.5-2.8. The eluate was collected in 1 mL fractions.
  • the pH of the protein fractions was immediately raised to neutral by addition of 100 ⁇ L of either 500 mM phosphate or Tris buffer, pH 7.6, to each 1 mL fraction. Variations of the reagent concentrations, labeling times, buffer composition, elution methods and other variables are possible that can yield equivalent results. Conjugates of the Fab fragment of goat anti-(rabbit Fc) and of protein G and protein A are prepared similarly.
  • Conjugates of the Fab fragment of goat anti-(mouse Fc) with the Alexa Fluor 488 succinimidyl ester were separately prepared, as described in Examples 4 and 5.
  • the conjugates had estimated degrees of substitution of ⁇ 1.9 (labeled as in Example 4) and ⁇ 3.0 (labeled as in Example 5), respectively, and virtually identical absorption and emission spectral maxima.
  • conjugates prepared using the fragment prepared as described in Example 5 were about 3.2-times more fluorescent than using the fragments that were prepared in Example 4 (FIG. 8) as detected by flow cytometry when bound to CD3 on Jurkat T cells. Similar results were observed with other dyes.
  • Native protein G has a high affinity binding (nanomolar) site for albumins, in particular ovalbumin. Equal weights of protein G and Texas Red ovalbumin (Molecular Probes. Inc.) were dissolved in PBS, pH 7.5. After one hour, the resulting complex was separated on a Sephacryl S-200 Superfine size-exclusion column and analyzed by SDS-PAGE and HPLC. Alternatively, the protein G is combined with a labeled albumin while the protein G is immobilized on any of the several immunoglobulins to which it binds, and the excess labeled albumin is washed away preceding elution of the albumin-labeled protein G complex from the matrix.
  • Submicrogram quantities of a target-binding antibody were complexed with submicrograms of a labeling protein in varying molar ratios of between about 1:1 and 1:20 to prepare an immunolabeling complex that was suitable for staining a sample.
  • mice monoclonal anti- ⁇ -tubulin in 1 ⁇ L PBS with 0.1% BSA was complexed with 0.5 ⁇ g of the Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse Fc) (prepared as in Example 4) or with 0.1 ⁇ g of the Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse Fc) (prepared as in Example 5) in 5 ⁇ L of PBS for 10 minutes at room temperature.
  • the immunolabeling complex can be used immediately for staining tubulin in fixed-cell preparations (Example 16) or any excess unbound Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse Fc) in the immunolabeling complex can be captured with non-immune mouse IgG (Example 9) for combination with other antibody conjugates, including those of targeting antibodies that have been directly conjugated to other labels. Rabbit antibodies were labeled similarly using labeled goat anti-(rabbit Fc). Labeling of targeting antibodies with a labeled protein A, protein L, protein G, protein G complexed with a labeled albumin, or other immunoglobulin-binding peptides or proteins proceeds similarly.
  • a labeled protein that is selective for the specific isotype of the primary antibody (e.g. anti-(mouse IgG 1 ) for a mouse IgG 1 isotype primary antibody).
  • a goat antibody that was selective for mouse IgG 1 isotype monoclonal antibodies, routine and optimal use for labeling unmatched mouse isotypes required greater amounts of immunolabeling complexes and was somewhat less reliable.
  • Immunolabeling complexes were prepared as described in Example 8. To the immunolabeling complex was added to each tube 25 ⁇ L of a 14.1 mg/mL stock solution of unlabeled mouse IgG to capture excess immunolabeling complexes. As shown in FIG. 1, not all of the immunoglobulin-binding protein was necessarily complexed with the target-binding antibody to form an immunolabeling complex. Consequently, particularly for applications in which labeling complexes of multiple primary antibodies from the same species (e.g. mouse monoclonal antibodies) or crossreacting species (e.g. mouse and human antibodies, FIG.
  • the most effective capturing component to capture excess immunoglobulin-binding protein is one that contains the binding site of the targeting agent.
  • whole mouse IgG or mouse serum was shown to be an effective and inexpensive reagent when the immunoglobulin-binding protein was bound to a segment of a mouse monoclonal antibody.
  • the mouse IgG was added in excess to the amount of immunoglobulin-binding protein and incubated for a period of approximately 1-5 minutes, or longer.
  • the immunolabeling complex It is preferred to prepare the immunolabeling complex and then add the capturing component shortly before the experiment. The rapid quenching effect permits this to be done within minutes of performing labeling of the sample by the immunolabeling complex. If desired, the excess capturing component can be removed following labeling of the sample by a simple wash step. Alternatively, fixation of the stained sample by aldehyde-based fixatives or other reagents or methods subsequent to incubation with the immunolabeling complex can provide permanent immobilization of the immunolabeling complex on its target in the sample. As an alternative to adding a soluble capturing component to the immunolabeling complex, the capturing component can be immobilized on an insoluble matrix such as agarose and the immunolabeling complex contacted with that matrix.
  • an insoluble matrix such as agarose
  • a preferred matrix when labeling mouse antibodies to mouse antigens is mouse IgG immobilized on agarose. Excess labeled anti-rabbit antibodies can be captured using rabbit IgG that is free in solution or immobilized. Alternatively, the immunolabeling complex can be separated from any capturing component by chromatographic or electrophoretic means.
  • Microplates were equilibrated overnight with IgG from a mouse or non-mouse species, and then further blocked with BSA. Variable amounts of the biotinylated Fab fragment of goat anti-(mouse Fc) were added to each well and allowed to bind. After washing, streptavidin-HRP and the Amplex Red peroxidase substrate were added. HRP activity was detected by the addition of H 2 O 2 using the Amplex Red Peroxidase Assay Kit (Molecular Probes, Inc., Eugene, Oreg.).
  • the goat anti-mouse IgG antibody reaction with mouse antibody was set at 100% and the crossreacting antibodies were expressed as a percentage compared the mouse on mouse data.
  • the data in Table 1 show that the Fab fragment of the goat anti-(mouse Fc) antibody of the current invention does not strongly bind to the goat or sheep Fc domain; however, one skilled in the art could generate antibodies that will react with the goat and sheep Fc domain or the Fc domain of any other species.
  • Biotinylated Fab goat anti-(mouse Fc) was used in this example because it provided a convenient method to quantitate the amount of crossreactivity in a conventional method but it could have been accomplished using a fluorophore Fab labeled goat anti-(mouse Fc).
  • mouse monoclonal anti-biotin (MW 145,000) in 8.0 ⁇ L PBS was added varying amounts of the Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse Fc) (MW ⁇ 50,000) (prepared as in Example 4) to form an immunolabeling complex. After equilibration for 20 min, a 100 ⁇ L aliquot was added to a 96-well microplate coated with biotinylated BSA. After 30 minutes, the plates were washed and the residual fluorescence was quantitated using a fluorescence microplate reader using excitation at 485+/ ⁇ 10 nm and detecting emission at 530+/ ⁇ 12.5 nm. As shown in FIG.
  • a molar ratio of the Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse Fc) to the anti-biotin between 5 to 20 was sufficient to form appreciably detectable complexes (FIG. 3; fluorescence quantitated, performed in triplicate (circles); control experiments performed but without adding the primary anti-biotin antibody (solid squares)).
  • a molar ratio of about 5 to about 10 was preferred for this pair of immunoglobulin-binding protein and target antibody. This ratio can be varied somewhat to increase or decrease the signal or to affect the consumption of valuable reagents.
  • the weight ratio of immunoglobulin-binding protein to target-binding antibody is particularly affected by the actual molecular weight of the immunoglobulin-binding protein.
  • a pre-equilibrated immunolabeling complex was prepared from 50 ⁇ g of an Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse Fc) and 15 ⁇ g of an anti-biotin monoclonal antibody (mAb).
  • the immunolabeling complex was rapidly diluted with capturing component sufficient to give a 6.2 molar excess over the anti-biotin mAb.
  • an aliquot was taken and added to a microplate well containing an excess of biotinylated BSA. After 30 minutes, the plates were washed and the remaining fluorescence was quantitated.
  • Displacement of the labeling protein from the target-binding antibody through exchange was measured by any time-dependent decrease in fluorescence in the microplate well.
  • the fragments prepared as described in Example 4 had 68 percent fragments bound to the target-binding antibody after 30 minutes compared to 87 percent of bound fragments that were prepared according to Example 5.
  • One hour showed a similar decrease, 56 percent and 68 percent respectively.
  • the labeling protein was shown to undergo a stable interaction with the target-binding antibody, with a lifetime for half exchange under these conditions of 3.5 hours.
  • Dissociation rates were measured for labeling protein prepared according to Example 4 and for labeling protein prepared according to Example 5, demonstrating the greater stability of immunolabeling complexes made using the labeling proteins prepared according to Example 5.
  • Culturable cells such as bovine pulmonary artery endothelial cells (BPAEC) were grown on a 22 ⁇ 22 mm glass coverslip. The cells were fixed for 10 minutes using 3.7% formaldehyde in DMEM with fetal calf serum (FCS) at 37° C. The fixed cells were washed 3 times with PBS. The cells were permeabilized for 10 min with 0.02% Triton X-100 in PBS, washed 3 ⁇ with PBS and blocked for 30 min with 1% BSA in PBS. Variations of the cell type and cell preparation, fixation, and permeabilization methods, including methods for antigen retrieval, are well known to scientists familiar with the art. An immunolabeling complex was prepared as described in Example 8.
  • BPAEC bovine pulmonary artery endothelial cells
  • the immunolabeling complex was added directly to the fixed and permeabilized cells in an amount sufficient to give a detectable signal if there is a binding site for the primary antibody present in the sample. After an incubation period that was typically 10-60 minutes (usually about 15-30 minutes), the cells were washed with fresh medium and the labeling was evaluated by methods suitable for detection of the label. Staining by the immunolabeling complex can be additionally preceded, followed by or combined with staining by additional reagents, such as DAPI, which yields blue-fluorescent nuclei.
  • additional reagents such as DAPI
  • Cells were fixed and permeabilized as described in Example 14. Multiple immunolabeling complexes were individually prepared from a variety of labeling proteins, according to the procedure described in Example 8. The multiple immunolabeling complexes were either used individually or sequentially to stain the cells, according to the procedure described in Example 14, or two or more immunolabeling complexes were formed then co-mixed in a single staining solution and used to simultaneously stain the sample. The optimal method for cell fixation and permeabilization and the best ratio for combination of the immunolabeling complexes are typically determined by preliminary experimentation using single immunolabeling complexes or multiple immunolabeling complexes used in combination.
  • a first immunolabeling complex was prepared from an Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse Fc) and mouse monoclonal anti- ⁇ -tubulin
  • a second immunolabeling complex was prepared from an Alexa Fluor 568 dye-labeled Fab fragment of goat anti-(mouse Fc) and mouse monoclonal anti-vimentin (anti-vimentin was an ascites fluid preparation)
  • a third immunolabeling complex was prepared from an Alexa Fluor 647 dye-labeled Fab fragment of goat anti-(mouse Fc) and mouse monoclonal anti-cdc6 peptide antibody (Molecular Probes).
  • the immunolabeling complexes that are used in combination do not have to be targeted toward antibodies from the same species.
  • complexes of Alexa Fluor 488 dye-labeled goat anti-(mouse IgG 1 Fc) with a mouse IgG 1 monoclonal target-binding antibody and an Alexa Fluor 594 dye-labeled goat anti-(rabbit Fc) with a rabbit primary target-binding antibody can be prepared and used in combined staining protocols.
  • a mouse intestine cryosection (University of Oregon histology core facility), a cross-section of about 16 ⁇ m thickness, was mounted on a slide. The intestine was perfused and fixed with 4% formaldehyde prior to dissection, embedding, and sectioning. The tissue section was rehydrated for 20 minutes in PBS. An immunolabeling complex was prepared as described in Example 8.
  • mouse monoclonal anti-cdc6 peptide (a nuclear antigen) in 1 ⁇ L PBS with 0.1% BSA was complexed with 0.5 ⁇ g of the Alexa Fluor 350 dye-labeled Fab fragment of goat anti-(mouse IgG 1 Fc) (prepared as in Example 4) in 5 ⁇ L of PBS for 10 minutes at room temperature.
  • Excess Fab fragment of goat anti-(mouse IgG 1 Fc) was captured with 25 ⁇ L of a 14.1 mg/mL stock of unlabeled mouse IgG.
  • the tissue was permeabilized with 0.1% Triton X-100 for 10 min.
  • tissue was washed two times with PBS and was blocked in 1% BSA for 30 min.
  • the immunolabeling complex was added directly to the tissue for 30 minutes and washed three times in PBS.
  • the sample was mounted in Molecular Probes' Prolong antifade mounting medium and observed by fluorescence microscopy using optical filters appropriate for the Alexa Fluor 350 dye. Results showed that the mouse monoclonal anti-cdc6 peptide immunolabeling complex showed specific nuclear labeling in the mouse intestine tissue section. Variations of the tissue type and tissue preparation, fixation and permeabilization methods, mounting methods, including methods for antigen retrieval, are well known to scientists familiar with the art.
  • Mouse brain cryosections were labeled with a pre-formed complex of horseradish peroxidase (HRP)-labeled goat anti-(mouse IgG 1 Fc) antibody and a mouse IgG 1 monoclonal anti-(glial fibrillary acidic protein (GFAP)) prepared essentially as in Example 8 using a molar ratio of labeling protein to monoclonal antibody of 3. Staining of the mouse tissues was essentially as in Example 16.
  • HRP horseradish peroxidase
  • GFAP mouse IgG 1 monoclonal anti-(glial fibrillary acidic protein
  • the staining localization and intensity was compared to that of (a) goat anti-mouse IgG HRP conjugate and mouse anti-GFAP, (b) the Alexa Fluor 488 dye-labeled Fab fragment of goat anti-mouse IgG 1 Fc) antibody complex of mouse anti-GFAP, (c) Alexa Fluor 488 goat anti-mouse IgG secondary antibody and mouse anti-GFAP, and (d) a direct conjugate of the Alexa Fluor 488 dye with mouse anti-GFAP.
  • the HRP-conjugated probes were incubated with Alexa Fluor 488 tyramide using TSA Kit #2 (Molecular Probes, Inc.) according to standard procedures.
  • tissue staining patterns in each case were similar and consistent with the expected staining pattern of mouse anti-GFAP and staining was essentially free of nonspecific background.
  • the relative fluorescence intensities of staining measured by digital imaging were sequentially: 541 relative intensity units for the HRP-goat anti-(mouse IgG 1 Fc) complex of mouse anti-GFAP and (using the combinations indicated by the letters above): (a) 539, (b) 234, (c) 294, and (d) 255 relative intensity units.
  • a first immunolabeling complex was prepared from an Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse IgG 1 Fc) and mouse monoclonal anti-(human CD8)
  • a second immunolabeling complex was prepared from an R-phycoerythrin-conjugated Fab fragment of goat anti-(mouse IgG 1 Fc) and mouse anti-(human CD3)
  • a third immunolabeling complex was prepared from an Alexa Fluor 647 dye-labeled Fab fragment of goat anti-(mouse IgG 1 Fc) and mouse anti-(human CD4).
  • the complexes were prepared as described in Example 8 and were each blocked with 20 ⁇ g (1.3 ⁇ L of 14.1 ⁇ g/mL) of mouse IgG for 10 minutes at room temperature.
  • the first immunolabeling complex was added to 100 ⁇ L of whole blood and incubated for 15 min.
  • the cells were washed with PBS and 280.5 ⁇ L of the second immunolabeling complex was added and incubated for 15 min.
  • the cells were again washed, and 46.2 ⁇ L of the third labeling complex was added and incubated for 15 min. After the final incubation, the red blood cells were lysed with cell-lysis buffer.
  • the cells were resuspended in 1% formaldehyde/PBS and analyzed on a FACS Vantage flow cytometer using a 488 nm argon-ion laser for excitation of the first and second immunolabeling complexes and a 633 nm red He—Ne laser for excitation of the third immunolabeling complex (FIGS. 5 a, 5 b ).
  • the emission band pass filters used for selective detection of the dyes are 525+/ ⁇ 10 nm for the Alexa Fluor 488 (CD8), 585+/ ⁇ 21 nm for R-PE (CD3) and 675+/ ⁇ 10 nm for the Alexa Fluor 647 dye (CD4).
  • FIG. 5 a and 5 b show that the instant invention can be used in a 3-color immunophenotyping experiment using peripheral blood lymphocytes.
  • CD3-positive T cells were stained with the R-phycoerythrin-conjugated Fab fragment of goat anti-(mouse Fc) and mouse anti-human CD3), upper left (UL) quadrant, FIG. 5 a.
  • CD4-positive cells, a T cell subset are identified using Alexa Fluor 647 dye-labeled Fab fragment of goat anti-(mouse IgG 1 Fc) and mouse anti-(human CD4), UL quadrant, FIG.
  • T cell subset a T cell subset, were identified using Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse IgG 1 Fc) and mouse monoclonal anti-(human CD8), lower right (LR) quadrant, FIG. 5 b.
  • Exposed antigens of live cells including cultured cells and cells from biological fluids such as blood and cerebrospinal fluid can be simultaneously or sequentially stained by combinations of immunolabeling complexes, including antibodies to the same target labeled with two or more separately detectable immunoglobulin-binding proteins.
  • a first immunolabeling complex was made by combining 2.5 ⁇ g Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse IgG 1 Fc) with 0.5 ⁇ g mouse anti-human CD3 (Caltag at 200 ⁇ g/mL), according to the procedure described in Example 4.
  • a second immunolabeling complex was made by combining 5.0 ⁇ g Alexa Fluor 647 dye-labeled Fab fragment of goat anti-(mouse IgG 1 Fc) with 0.5 ⁇ g mouse anti-human CD3, according to the procedure in Example 4.
  • Each complex was separately incubated at room temperature for 5 minutes, and each complex was then separately combined with an excess of mouse IgG (14.1 mg/mL) for 5 min at room temperature to capture excess unbound dye-labeled Fab fragments.
  • the two immunolabeling complexes were then added in different percentage combinations (see Table 2) to 100 ⁇ L of washed heparinized blood.
  • the cells were incubated with the respective combinations of complexes for 20 min on ice.
  • the red blood cells were then lysed with a cell-lysis buffer.
  • the cells were resuspended in 1% formaldehyde/PBS and analyzed on a FacVantage flow cytometer using a 488 nm argon 633 HeNe laser for excitation and a 530+/ ⁇ 10 nm band pass emission filter (FL1), and a 640 long pass filter (FL4).
  • Five samples of different combined percentages (Table 2) were compared by flow cytometry, with signals being collected in FL1 and FL4.
  • the FL1 and FL4 intensities for each percentage combination were normalized by dividing the FL1 and FL4 channel intensities for such combinations by the intensities of the 100% Alexa Fluor 488 dye- and 100% Alexa Fluor 647 dye-labeled cells, respectively.
  • Bovine heart mitochondria were isolated (Hanson et al., Electrophoresis 22, 950 (2001)). The isolated mitochondria were resuspended to ⁇ 10 mg/mL in 100 mM Tris-HCl, pH 7.8, 1 mM phenylmethylsulfonyl fluoride (a protease inhibitor), 2% SDS and insoluble material was removed by centrifugation for 10 minutes at 10,000 ⁇ g in a tabletop centrifuge. The protein concentration of the lysate was checked by the BCA assay (Pierce, Rockford, Ill.).
  • Samples for gel electrophoresis were prepared by mixing lysate, water, and loading buffer to the appropriate concentrations (final concentration of loading buffer in samples: 58 mM Tris/HCl, 10% glycerol, 2% SDS, 0.02 mg/mL bromphenol blue, 50 mM DTT, pH 8.6). The samples were then heated to 90° C. for 5 minutes before loading on the gel and separated on a 13% SDS-PAGE gel. Two-fold serial dilution of the extracts ranging from 8 ⁇ g of extract down to 0.03 ⁇ g were loaded on the SDS-PAGE gel. The proteins were transferred to PVDF membrane for 1.5 hours using a semi-dry transfer system according to manufacturer's directions (The W.E.P. Company, Concord, Calif.). The PVDF membrane was blocked for l hour in 5% milk.
  • Immunolabeling complexes were made with mouse monoclonal antibodies that recognize two different mitochondrial proteins. Alexa Fluor 647 dye-labeled Fab fragment of goat anti-(mouse IgG 1 Fc) (5 ⁇ L of a 1 mg/mL stock, prepared as in Example 4) was incubated with 21 ⁇ L (0.88 mg/mL) mouse anti-(CV-alpha) and Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse IgG 1 Fc) (5 ⁇ L of a 1 mg/mL stock, prepared as in Example 4) was incubated with 19 ⁇ L (0.88 mg/mL) mouse anti-(CIII-core2) (Molecular Probes, Eugene, Oreg.).
  • Hybridoma supernatant is harvested and added to the wells. If the hybridoma are producing the desired antibody, i.e. antibodies that bind to the labeled antigen, polarization of the florescence corresponding to the labeled antigen will allow visualization of those wells containing antigen specific antibody.
  • the amount of IgG which the hybridomas produce can be simultaneously identified by polarization of the fluorescence corresponding to the labeled Fab fragments. This method thus allows for both quantitation of the amount of antibody present in a specific amount of hybridoma supernatant and the affinity of the monoclonal antibodies for the antigen.
  • the reagents employed in the preceding examples are commercially available or can be prepared using commercially available instrumentation, methods, or reagents known in the art or whose preparation is described in the examples. It is evident from the above description and results that the subject invention is greatly superior to the presently available methods for determining the presence of a target in a biological sample.
  • the subject invention overcomes the shortcomings of the currently used methods by allowing small quantities of antibodies to be labeled and in unlimited media while maintaining specificity and sensitivity.
  • the examples are not intended to provide an exhaustive description of the many different embodiments of the invention.

Abstract

The present invention provides novel immunolabeling complexes and certain components of such complexes, as well as methods of preparing and using such complexes, and kits for use in preparing labeling proteins and for immunolabeling. The pre-formed immunolabeling complexes of the invention comprise both a target-binding antibody and a labeling protein that contains covalently attached labels, where the labeling protein binds selectively and with high affinity to a selected region of the target-binding antibody. Novel labeling proteins of the invention include non-antibody peptides and proteins, such as a complex of protein G and a labeled albumin, and monovalent antibody fragments, such as labeled Fab fragments of an anti-Fc antibody. In methods of the invention, the preformed immunolabeling complexes are added to the sample alone or in combination, for purposes of labeling and optionally detecting the target of interest.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to provisional applications, serial No. 60/329,068, filed Oct. 2, 2001 and provisional application filed Apr. 1, 2002, which disclosures are herein incorporated by reference.[0001]
  • FIELD OF INVENTION
  • The present invention relates to novel immunolabeling compositions and methods for use in the detection and measurement of one or more targets in a biological sample. The invention has applications in the fields of molecular biology, cell biology screening, immunohistochemistry, diagnostics, and therapeutics. [0002]
  • BACKGROUND OF THE INVENTION
  • Immunolabeling is a method for qualitative or quantitative determination of the presence of a target in a sample, wherein antibodies are utilized for their specific binding capacity. The antibodies form a complex with the target (antigen), wherein a detectable label is present on the antibody or on a secondary antibody. The detectable label is a key feature of immunolabeling, which can be detected directly or indirectly. The label provides a measurable signal by which the binding reaction is monitored providing a qualitative and/or quantitative measure of the degree of binding. The relative quantity and location of signal generated by the labeled antibodies can serve to indicate the location and/or concentration of the target. The label can also be used to select and isolate labeled targets, such as by flow sorting or using magnetic separation media. Examples of labels include but are not limited to radioactive nucleotides ([0003] 125I, 3H, 14C, 32P), chemiluminescent, fluorescent, or phosphorescent compounds (e.g., dioxetanes, xanthene, or carbocyanine dyes, lanthanide chelates), particles (e.g., gold clusters, colloidal gold, microspheres, quantum dots), and enzymes (e.g., peroxidases, glycosidases, phosphatases, kinases). Ideally, the label is attached to the antibody in a manner that does not perturb the antibody's binding characteristics but enables the label to be measured by an appropriate detection technology. The choice of labels is influenced by factors such as ease and sensitivity of detection, equipment availability, background in the sample (including other labels) and the degree to which such labels are readily attached to the particular antibody. Both direct and indirect labeling of antibodies are utilized for immunolabeling. Direct labeling utilizes only a primary antibody, i.e. the antibody specific for the target, bound to the label. In contrast, indirect labeling utilizes a secondary antibody bound to the label, which is specific for the primary antibody, e.g. a goat anti-rabbit antibody. The principal differences in immunolabeling methods and materials reside in the way that the label is attached to the antibody-antigen complex, the type of label that is used, and the means by which the antibody-antigen complex is detected.
  • Limitations for direct labeling primary antibodies include the need for buffers free of primary amines, or carrier proteins such as bovine serum albumin (BSA), and other compounds such as tris-(hydroxymethyl)aminomethane (TRIS), glycine, and ammonium ions. These materials are, however, common components in antibody buffers and purification methods, and it may not be possible or feasible to remove them prior to the coupling reaction. In particular, many monoclonal antibodies are available only as ascites fluid or in hybridoma culture supernatants, or diluted with carrier proteins, such as albumins. Thus, direct labeling of antibodies in ascites fluid or other medias containing interfering compounds is not attainable. [0004]
  • The indirect immunolabeling method typically involves a multi-step process in which an unlabeled first antibody (typically a primary antibody) is directly added to the sample to form a complex with the antigen in the sample. Subsequently, a labeled secondary antibody, specific for the primary antibody, is added to the sample, where it attaches noncovalently to the primary antibody-antigen complex. Alternatively, a detectable label is covalently attached to an immunoglobulin-binding protein such as protein A and protein G to detect the antibody-antigen complex that has previously been formed with the target in the sample. Using ligands, such as streptavidin, that are meant to amplify the detectable signal also expands this cascade binding. [0005]
  • Indirect immunolabeling often results in false positives and high background. This is due to the fact that secondary antibodies, even when purified by adsorption against related species, nevertheless can exhibit significant residual cross-reactivity when used in the same sample. For example, when mouse tissue is probed with a mouse monoclonal antibody, the secondary antibody must necessarily be a labeled anti-mouse antibody. This anti-mouse antibody will detect the antibody of interest but will inevitably and additionally detect irrelevant, endogenous mouse immunoglobulins inherent in mouse tissue. This causes a significant background problem, especially in diseased tissues, which reduces the usefulness and sensitivity of the assay. Thus, the simultaneous detection of more than one primary antibody in a sample without this significant background interference depends on the availability of secondary antibodies that 1) do not cross-react with proteins intrinsic to the sample being examined, 2) recognize only one of the primary antibodies, and 3) do not recognize each other (Brelje, et al., METHODS IN CELL BIOLOGY 38,97-181, especially 111-118 (1993)). [0006]
  • To address the background problem in indirect labeling, a number of strategies have been developed to block access of the anti-mouse secondary antibodies to the endogenous mouse immunoglobulins. One such strategy for blocking involves complexing the primary antibody with a selected biotinylated secondary antibody to produce a complex of the primary and secondary antibodies, which is then mixed with diluted normal murine serum (Trojanowski et al., U.S. Pat. No. 5,281,521 (1994)). This method is limited by the necessity to utilize an appropriate ratio of primary-secondary complex. Too low a ratio of primary-secondary complex will cause a decrease in specific staining and increased background levels due to the uncomplexed secondary anti-mouse antibody binding to endogenous mouse antibodies. However, the ability of a whole IgG antibody (as was used in the referenced method) to simultaneously bind and crosslink two antigens results in too high a ratio, causing the complex to precipitate or form complexes that are too large to penetrate into the cell or tissue. [0007]
  • Another strategy for blocking access to endogenous immunoglobulins in the sample involves pre-incubating the sample with a monovalent antibody, such as Fab′ fragments, from an irrelevant species that recognize endogenous immunoglobulins. This approach requires large quantities of expensive Fab′ fragments and gives mixed results and adds at least two steps (block and wash) to the overall staining procedure. The addition of a cross-linking reagent has resulted in improved reduction of background levels (Tsao, et al., U.S. Pat. No. 5,869,274 (1997)) but this is problematic when used with fluorophore-labeled antibodies. The cross-linking causes an increase in the levels of autofluorescence (J. Neurosci. Meth. 83, 97 (1998); Mosiman et al., Methods 77, 191 (1997); Commun. Clin. Cytometry 30, 151 (1997); Beisker et al., Cytometry 8, 235 (1987)) and thus the background. In addition, pre-incubation with a cross-linking reagent often masks or prevents the antibody from binding to its antigen (J. Histochem. Cytochem. 45, 327 (1997); J. Histochem. Cytochem. 39, 741 (1991); J. Histochem. Cytochem. 43, 193 (1995); Appl. Immunohistochem. Molecul. Morphol. 9, 176 (2001)). [0008]
  • In a variation of this blocking strategy, a multi-step sequential-labeling procedure is used to overcome the problems of cross-reactivity. The sample is incubated with a first antibody to form a complex with the first antigen, followed by incubation of the sample with a fluorophore-labeled goat Fab anti-mouse IgG to label the first antibody and block it from subsequently complexing when the second antibody is added. In the third step, a second mouse antibody forms a complex with the second antigen. Being blocked from cross-reacting with the first antibody, the second mouse antibody is detected with a standard indirect-labeling method using a goat anti-mouse antibody conjugated to a different fluorescent dye (J. Histochem. Cytochem. 34, 703 (1986)). This process is complex in that it requires multiple incubation steps and washing steps and it still cannot be used with mouse antibodies to probe mouse tissue. [0009]
  • Another blocking method is disclosed in the animal research kit (ARK) developed by DAKO. In this kit, a primary antibody is complexed with biotin-labeled goat Fab anti-mouse IgG and excess free Fab is blocked with normal mouse serum. However, since the Fab used in this process is generated from the intact IgG (rather than a selected region) there is a potential for the formation of anti-paratope or anti-idiotype antibodies that will block the antigen-binding site and prevent immunolabeling. The biotinylated antibody also requires subsequent addition of a labeled avidin or streptavidin conjugate for its subsequent visualization. [0010]
  • The present invention is advantageous over previously described methods and compositions in that it provides the benefits of indirect labeling with the easy and flexibility of direct labeling for determination of a desired target in a biological sample. The present invention provides labeled monovalent proteins specific for a target-binding antibody, which are complexed prior to addition with a biological sample. Because these monovalent proteins are not bivalent antibodies, precipitation and cross-linking are not a problem. Therefore the compositions of the present invention can be used with immunologically similar monoclonal or polyclonal antibodies of either an identical isotype or different isotypes. The monovalent labeling proteins are specific for the Fc region of target-binding antibodies, these proteins will not interfere with the binding region of the primary antibody. In addition, the monovalent labeling proteins are not negatively affected by the presence of primary amines like BSA, gelatin, hybridoma culture supernatants or ascites fluid, thus primary antibodies present in these media can be effectively labeled with the labeling proteins of the present invention. Thus, the present invention provides numerous advantages over the conventional methods of immunolabeling. [0011]
  • SUMMARY OF THE INVENTION
  • Methods and compositions are provided for determining the presence, absence or location of a desired target in a biological sample. The methods initially involve pre-forming the immunolabeling complex, the target-binding antibody and the labeling protein, followed by addition to a biological sample and determination of the desired target. The methods of the invention are broadly applicable to immunolabeling any antigen target in any sample from any origin.[0012]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a schematic representation of the formation of the immunolabeling complex (target-bindingprotein and labeling protein). [0013]
  • FIG. 2 graphically represents species specificity of goat Fab anti-(mouse Fc), as observed using a microplate coated with IgG of various species. The various species were blocked with BSA, reacted with biotinylated goat Fab anti-(mouse Fc), washed, and then treated with streptavidin-horseradish peroxidase (HRP), followed by hydrogen peroxide (H[0014] 2O2) and the Amplex Red peroxidase detection reagent.
  • FIG. 3 graphically depicts the optimal molar ratio of a goat Fab anti-(mouse Fc) protein. Varying amounts of an [0015] Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse Fc) were added to a constant amount of anti-biotin monoclonal antibody (mAb). This mixture was equilibrated for 20 minutes, and then added to biotinylated-BSA in a microplate well. After allowing time to bind, the plates were washed and the remaining fluorescence was quantitated. The analysis was performed in triplicate (circles). Control experiments were performed, as described above, but without adding the primary anti-biotin antibody (solid squares).
  • FIG. 4 illustrates a comparison of the fluorescence intensity (Example 6) for labeling protein prepared in homogeneous solution (Example 4) and labeling protein prepared on a column (Example 5). [0016]
  • FIG. 5A: CD3-positive R-phycoerythrin (R-PE) stained T cells are shown in the upper left (UL) and upper right (UR) quadrants (Example 18). The relative percentages of total lymphocytes that are CD3-positive cells are 83.3% (UL+UR). The relative percentage of CD8-[0017] positive Alexa Fluor 488 dye-stained lymphocytes and CD3-positive R-PE dye-stained lymphocytes is 35.1% (UR quadrant). The lower left quadrant (LL, 20.4%) shows CD3-negative lymphocytes (i.e. non-T cells) comprised of NK cells, B cells and some monocytes. In the lower right (LR, 2.7%) region are non-T cells, which are nonspecifically stained.
  • FIG. 5B: CD3-positive T-cells are subdivided into [0018] Alexa Fluor 647 CD4-positive and Alexa Fluor 488 CD8-positive (Example 18). CD4-positive cells represent 50.9% of total lymphocytes (UL quadrant) and CD8-positive cells represent 24.5% of the total lymphocytes (LR quadrant). The 23.1% of cells in the LL quadrant are non-T cells, while the 1.5% of cells in UR quadrant are likely nonspecifically stained lymphocytes.
  • FIG. 6 illustrates high-performance size-exclusion chromatographic analysis of [0019] Alexa Fluor 488 dye-labeled goat Fab anti-(mouse Fc) labeling protein binding to a mouse IgG1 target-binding antibody. The labeling protein, alone, appears as a peak at 38 minutes; the target-binding antibody, alone, appears as a peak at 33 minutes. When labeling protein and target-binding antibody are mixed together at a molar ratio of ˜5:1 (labeling protein:target-binding antibody), the resulting immunolabeling complex appears as a peak at 29 minutes (Example 10).
  • FIG. 7 is a cartoon depiction of the process for preparing improved labeling proteins.[0020]
  • DESCRIPTION OF THE SPECIFIC EMBODIMENTS
  • I. Definitions [0021]
  • To assist in the understanding of the invention, the following terms, as used herein, are defined below. [0022]
  • “Affinity” is defined as the strength of the binding interaction of two molecules, such as an antigen and its antibody, which is defined for antibodies and other molecules with more than one binding site as the strength of binding of the ligand at one specified binding site. Although the noncovalent attachment of a ligand to antibody is typically not as strong as a covalent attachment, “High affinity” is for a ligand that binds to an antibody having an affinity constant (K[0023] a) of greater than 104 M−1, typically 105-1011 M−1; as determined by inhibition ELISA or an equivalent affinity determined by comparable techniques such as, for example, Scatchard plots or using Kd/dissociation constant, which is the reciprocal of the Ka, etc.
  • “Antibody” is a protein of the immunoglobulin (Ig) superfamily that binds noncovalently to certain substances (e.g. antigens and immunogens) to form an antibody-antigen complex, including but not limited to antibodies produced by hybridoma cell lines, by immunization to elicit a polyclonal antibody response, by chemical synthesis, and by recombinant host cells that have been transformed with an expression vector that encodes the antibody. In humans, the immunoglobulin antibodies are classified as IgA, IgD, IgE, IgG, and IgM and members of each class are said to have the same isotype. Human IgA and IgG isotypes are further subdivided into subtypes IgA[0024] 1, and IgA2, and IgG1, IgG2, IgG3, and IgG4. Mice have generally the same isotypes as humans, but the IgG isotype is subdivided into IgG1, IgG2a, IgG2b, and IgG3 subtypes. Thus, it will be understood that the term “antibody” as used herein includes within its scope (a) any of the various classes or sub-classes of immunoglobulin, e.g., IgG, IgM, IgE derived from any of the animals conventionally used and (b) polyclonal and monoclonal antibodies, such as murine, chimeric, or humanized antibodies. Antibody molecules have regions of amino acid sequences that can act as an antigenic determinant, e.g. the Fc region, the kappa light chain, the lambda light chain, the hinge region, etc. An antibody that is generated against a selected region is designated anti-[region], e.g. anti-Fc, anti-kappa light chain, anti-lambda light chain, etc. An antibody is typically generated against an antigen by immunizing an organism with a macromolecule to initiate lymphocyte activation to express the immunoglobulin protein. The term antibody, as used herein, also covers any polypeptide or protein having a binding domain that is, or is homologous to, an antibody binding domain, including, without limitation, single-chain Fv molecules (scFv), wherein a VH domain and a VL domain are linked by a peptide linker that allows the two domains to associate to form an antigen binding site (Bird et al., Science 242, 423 (1988) and Huston et al., Proc. Natl. Acad. Sci. USA 85, 5879 (1988)). These can be derived from natural sources, or they may be partly or wholly synthetically produced.
  • “Antibody fragments” refers to fragments of antibodies that retain the principal selective binding characteristics of the whole antibody. Particular fragments are well-known in the art, for example, Fab, Fab′, and F(ab′)[0025] 2, which are obtained by digestion with various proteases and which lack the Fc fragment of an intact antibody or the so-called “half-molecule” fragments obtained by reductive cleavage of the disulfide bonds connecting the heavy chain components in the intact antibody. Such fragments also include isolated fragments consisting of the light-chain-variable region, “Fv” fragments consisting of the variable regions of the heavy and light chains, and recombinant single chain polypeptide molecules in which light and heavy variable regions are connected by a peptide linker. Other examples of binding fragments include (i) the Fd fragment, consisting of the VH and CH1 domains; (ii) the dAb fragment (Ward, et al., Nature 341, 544 (1989)), which consists of a VH domain; (iii) isolated CDR regions; and (iv) single-chain Fv molecules (scFv) described above. In addition, arbitrary fragments can be made using recombinant technology that retains antigen-recognition characteristics.
  • “Antigen” means a molecule that induces, or is capable of inducing, the formation of an antibody or to which an antibody binds selectively, including but not limited to a biological material. Antigen also refers to “immunogen”. An antibody binds selectively to an antigen when there is a relative lack of cross-reactivity with or interference by other substances present. [0026]
  • “Biological material” means a material of biological origin or a synthetic variant of a material of biological origin, including but not limited to cells, bodily fluids, membranes, proteins, amino acids, nucleic acids, carbohydrates, lipids, and any polymers or combinations thereof, including natural as well as synthetic polymers that are optionally relatively homogeneous or heterogeneous. [0027]
  • “Biological Sample” refers to any material that may contain a target to which an antibody can bind. Typically, the sample comprises tissue, cell or cells, cell extracts, cell homogenates, purified or reconstituted proteins, recombinant proteins, bodily and other biological fluids, viruses or viral particles, prions, subcellular components, or synthesized proteins. Possible sources of cellular material used to prepare the sample of the invention include without limitation plants, animals, fungi, protists, bacteria, archae, or cell lines derived from such organisms. [0028]
  • “Biotin” means any biotin derivative, including without limitation, substituted and unsubstituted biotin, and analogs and derivatives thereof, as well as substituted and unsubstituted derivatives of caproylamidobiotin, biocytin, desthiobiotin, desthiobiocytin, iminobiotin, and biotin sulfone. [0029]
  • “Biotin-binding protein” means any protein that binds selectively and with high affinity to biotin, including without limitation, substituted or unsubstituted avidin, and analogs and derivatives thereof, as well as substituted and unsubstituted derivatives of streptavidin, ferritin avidin, nitroavidin, nitrostreptavidin, and Neutravidin™ avidin (a de-glycosylated modified avidin having an isoelectric point near neutral). [0030]
  • “Bodily or body fluid” means a fluid substance from the body, including but not limited to, for example, circulating blood and lymph, the chyle, the gastric, pancreatic, and intestinal juices, the saliva, bile, urine, aqueous humor, spinal fluid, muscle serum and ascites. [0031]
  • “Buffer” means a system that acts to minimize the change in concentration of a specific chemical species in solution against addition or depletion of this species. [0032]
  • “Cell or cells” means an autonomous self-replicating unit composed of protoplasm delimited by a cell membrane that may constitute an organism (in the case of unicellular organisms) or be a subunit of multicellular organisms (in which individual cells may be more or less specialized differentiated) for particular functions or plant cells. Cell or cells as used in the instant application also includes cells in cell culture medium, single cells, isolated cells, single-cell organisms; and “portions thereof” including, but not limited to, cell extracts, cell homogenates, cell lysates, and subcellular components. [0033]
  • “Complex” means two or more molecules held together by noncovalent bonding, which are typically noncovalent combinations of biomolecules such as a protein complexed with another protein. In contrast, a protein is covalently labeled with a substance when there is a covalent chemical bond between the substance and the protein. [0034]
  • “Detectable response” means a change in, or occurrence of, a signal that is detectable either by observation or instrumentally. Typically the detectable response is an optical response resulting in a change in the wavelength distribution patterns or intensity of absorbance or fluorescence or a change in light scatter, fluorescence lifetime, fluorescence polarization, or a combination of the above parameters. Other detectable responses include, for example, chemiluminescence, phosphorescence, radiation from radioisotopes, magnetic attraction, and electron density. [0035]
  • “Detectably distinct” means the signal is distinguishable or separable by a physical property either by observation or instrumentally. For example, but not limitation, a fluorophore is readily distinguishable, either by spectral characteristics or by fluorescence intensity, lifetime, polarization or photo-bleaching rate from another fluorophore in the sample, as well as from additional materials that are optionally present. [0036]
  • “Directly detectable” means that the presence of a material or the signal generated from the material is immediately detectable by observation, instrumentation, or film without requiring chemical modifications or additional substances. [0037]
  • “Drug-labeled protein” means a labeling protein of the invention, where instead of a detectable label being attached to the protein, a therapeutic agent or drug is attached. The term drug-labeled protein is used interchangeably with immunoconjugate. [0038]
  • “Enzyme” means a protein molecule produced by living organisms, or through chemical modification of a natural protein molecule, that catalyses chemical reactions of other substances without itself being destroyed or altered upon completion of the reactions. Examples of other substances, include but are not limited to, chemiluminescent, chromogenic, and fluorogenic substances. [0039]
  • “Immunoconjugates” are labeling proteins of the invention, where instead of a detectable label being attached to the protein, a therapeutic agent or drug is attached. The term immunoconjugate is used interchangeably with drug-labeled protein. [0040]
  • “Immunolabeling complex” means an antibody-protein complex made detectable, traceable, or therapeutically useful by incorporation of a detectable label, drug, or therapeutic into the protein that is noncovalently and selectively bound to the antibody. [0041]
  • “Kit” means a packaged set of related components, typically one or more compounds or compositions. [0042]
  • “Label” means a chemical used to facilitate identification and/or quantitation of a target substance. Illustrative labels include labels that can be directly observed or measured or indirectly observed or measured. Such labels include, but are not limited to, radiolabels that can be measured with radiation-counting devices; pigments, dyes or other chromogens that can be visually observed or measured with a spectrophotometer; spin labels that can be measured with a spin label analyzer; and fluorescent moieties, where the output signal is generated by the excitation of a suitable molecular adduct and that can be visualized by excitation with light that is absorbed by the dye or can be measured with standard fluorometers or imaging systems, for example. The label can be a luminescent substance such as a phosphor or fluorogen; a bioluminescent substance; a chemiluminescent substance, where the output signal is generated by chemical modification of the signal compound; a metal-containing substance; or an enzyme, where there occurs an enzyme-dependent secondary generation of signal, such as the formation of a colored product from a colorless substrate. The label may also take the form of a chemical or biochemical, or an inert particle, including but not limited to colloidal gold, microspheres, quantum dots, or inorganic crystals such as nanocrystals or phosphors (see, e.g., Beverloo, et al., Anal. Biochem. 203, 326-34 (1992)). The term label can also refer to a “tag” or hapten that can bind selectively to a labeled molecule such that the labeled molecule, when added subsequently, is used to generate a detectable signal. For instance, one can use biotin, iminobiotin or desthiobiotin as a tag and then use an avidin or streptavidin conjugate of horseradish peroxidase (HRP) to bind to the tag, and then use a chromogenic substrate (e.g., tetramethylbenzidine) or a fluorogenic substrate such as Amplex Red or Amplex Gold (Molecular Probes, Inc.) to detect the presence of HRP. In a similar fashion, the tag can be a hapten or antigen (e.g., digoxigenin), and an enzymatically, fluorescently, or radioactively labeled antibody can be used to bind to the tag. Numerous labels are known by those of skill in the art and include, but are not limited to, particles, fluorescent dyes, haptens, enzymes and their chromogenic, fluorogenic, and chemiluminescent substrates, and other labels that are described in the MOLECULAR PROBES HANDBOOK OF FLUORESCENT PROBES AND RESEARCH CHEMICALS by Richard P. Haugland, 6[0043] th Ed., (1996), and its subsequent 7th edition and 8th edition updates issued on CD Rom in November 1999 and May 2001, respectively, the contents of which are incorporated by reference, and in other published sources.
  • “Monovalent antibody fragment” refers to an antibody fragment that has only one antigen-binding site. Examples of monovalent antibody fragments include, but are not limited to, Fab fragments (no hinge region), Fab′ fragments (monovalent fragments that contain a heavy chain hinge region), and single-chain fragment variable (ScFv) proteins. [0044]
  • “Non-antibody immunoglobulin-binding protein” means a protein that binds selectively and noncovalently to a member of the Ig superfamily of proteins, including but not limited to proteins A, G, and L, hybrids thereof (A/G), recombinant versions and cloned versions thereof, fusions of these proteins with detectable protein labels, and lectins but the protein itself is not an antibody or an antibody fragment. [0045]
  • “Non-antibody immunoglobulin-binding peptide” means a peptide that selectively and noncovalently binds to a member of the Ig superfamily of proteins, such as a peptide that is selected by a process of screening against a biomolecular library. [0046]
  • “Peptide” means a polymer of amino acids, and includes polypeptides. [0047]
  • “Protein G” means proteins comprising one or more natural IgG-binding domains of protein G, hybrids of the natural IgG-binding domains, and mutants thereof wherein the variant retains the capability of binding IgG, or fragments thereof. [0048]
  • “Protein A” means proteins comprising one or more natural IgG-binding domains of protein A, hybrids of the natural IgG-binding domains, and mutants thereof wherein the variant retains the capability of binding IgG, or fragments thereof. [0049]
  • “Protein L” means proteins comprising one or more natural antibody light-chain-binding domains of protein L, hybrids of the natural antibody light-chain-binding domains, and mutants thereof wherein the variant retains the capability of binding antibody light chain, or fragments thereof. [0050]
  • “Selectively binds” refers to the situation in which one member of a specific intra or inter species binding pair will not show any significant binding to molecules other than its specific intra- or inter-species binding partner (e.g., an affinity of about 100-fold less), i.e. minimal cross-reactivity. [0051]
  • “Subcellular component” means functional units within a cell, including but not limited to, for example, membranes, organelles, receptors, nucleus, chromosomes, chloroplasts, peroxisomes, polyribosomes, mitochondria, lysosomes, DNA, RNA, endogenous proteins, peptides, enzymes, hormones, and factors. [0052]
  • “Target” means any entity to be detected by the association of the target-binding antibody with it. [0053]
  • II. Methods and Compositions [0054]
  • In accordance with the subject invention, methods and compositions are provided that facilitate the determination of a desired target in a biological sample. The method is performed in two steps. The immunolabeling complex, which contains the target-binding antibody and the labeling protein bound by a detectable label, is pre-formed followed by the addition to a sample suspected of containing the desired target. The labeling protein is either a monovalent Fab, fragment or a non-immunoglobulin peptide or protein, and specifically binds a selected region of the target-binding antibody. The labeling protein is covalently attached to one or more detectable labels, wherein the detectable labels can be the same or different allowing for multiparameter applications. After addition of the pre-formed immunolabeling complex to a sample, followed by sufficient time for the complex to bind with the target, detection of the label is determined. Methods of visualizing the label depend on the label attached to the labeling protein. [0055]
  • The labeling proteins bind selectively and with high affinity to a selected region of the target-binding antibody. These labeling proteins bind selectively to the same region of the target-binding antibody. The binding region for the labeling proteins may be a selected peptide linker (including the J region), light chain or heavy chain of the target-binding antibody; preferably the labeling protein binds the Fc region of the target-binding antibody. The target-binding antibody may be bound with one or more labeling proteins and the labeling proteins may be bound with one label, two similar labels, two distinct labels or multiple combinations thereof. Binding of more than one label, such as two different dyes on the same protein or on different proteins, used in different proportions, permits combinatorial applications that increase the number of detectable targets in the sample. Typically the same dye is used on a given labeling protein and the labeling proteins are monovalent Fab fragments or non-antibody peptide or proteins. [0056]
  • In this method, the labeling protein can be either an antibody fragment, such as, but not limited to, anti-Fc, an anti-Fc isotype, anti-J chain, anti-kappa light chain, anti-lambda light chain, or a single-chain fragment variable protein; or a non-antibody peptide or protein, such as, for example but not limited to, soluble Fc receptor, protein G, protein A, protein L, lectins, or a fragment thereof. Typically, the labeling protein is a Fab fragment specific to the Fc portion of the target-binding antibody or to an isotype of the Fc portion of the target-binding antibody. The monovalent Fab fragments are produced from either murine monoclonal antibodies or polyclonal antibodies generated in a variety of animals, for example but not limited to, rabbit or goat. These fragments can be generated from any isotype such as murine IgM, IgG[0057] 1, IgG2a, IgG2b or IgG3. Alternatively, a non-antibody protein or peptide such as protein G, or other suitable proteins, can be used alone or coupled with albumin wherein albumin is complexed with the detectable label. Preferred albumins of the invention include human and bovine serum albumins or ovalbumin.
  • Labels that are covalently attached to the labeling proteins are known by those of skill in the art and include, but are not limited to, radiolabels, pigments, dyes or other chromogens, spin labels, fluorescent compounds, haptens, electron transfer agents, and particles. The label can also be a precursor to a luminescent substance, a bioluminescent substance, a chemiluminescent substance, or a metal-containing substance. Preferred labels are fluorescent moieties such as those described in the MOLECULAR PROBES HANDBOOK OF FLUORESCENT PROBES AND RESEARCH CHEMICALS by R. P. [0058] Haugland 8th Ed., CD-ROM (2001). Particullary preferred are xanthenes (e.g., fluoreceins, rhodamines and rhodols) especially sulfonated xanthenes such as those disclosed in U.S. Pat. No. 6,130,101 and UK 9611997.9 and fluorinated xanthenes such as those disclosed in U.S. Pat. No. 6,162,931; and coumarins, especially sulfonated coumarins such as those described in U.S. Pat. No. 5,969,157 and fluorinated coumarins such as those disclosed in U.S. Pat. No. 5,830,912; and cyanines especially sulfonated cyanines disclosed and PCT/01/30404 publication. Other preferred labels are fluorescent proteins, especially phycobiliprotein and tandem-dye conjugates thereof as well as haptens such as biotin including desthibiotin) and enzymes, described in greater detail below.
  • Preparation of labeling proteins using low molecular weight reactive dyes is known by those of skill in the art and is well documented, e.g., by Richard P. Haugland, MOLECULAR PROBES HANDBOOK OF FLUORESCENT PROBES AND RESEARCH CHEMICALS, Chapters 1-3 (1996) and by Brinkley, Bioconjugate Chem. 3, 2 (1992). Labeling proteins typically result from mixing appropriate reactive dyes and the protein to be conjugated in a suitable solvent in which both are soluble. The majority of the preferred dyes of the invention are readily soluble in aqueous solutions, facilitating conjugation reactions with most biological materials. For those reactive dyes that are photoactivated, conjugation requires illumination of the reaction mixture to activate the reactive dye. [0059]
  • A particular advantage of the current invention over other methods of labeling target-binding antibodies is that it provides for a novel method to directly label the antibodies in any solution containing primary amines or non-antibody proteins, for example from ascites fluid, hybridoma culture supernatant, serum, in the presence of stabilizing proteins like BSA, gelatin, ammonium ions, or buffers like TRIS. Consequently, the methods of the instant invention provide a means to rapidly screen hybridomas for antibody expression and quality of antibody properties (e.g. affinity, specificity, cross-reactivity, isotype, abundance). [0060]
  • In the methods described above, the labeling complexes, whether for single or multicolor detection systems, are combined with a sample thought to contain target materials in any way that facilitates contact between the immunolabeling complexes and the target of interest. Preferred targets of the invention are biological materials and include, for example, but not limited to, a cell, receptor, membrane, protein, nucleic acid, or carbohydrate, but can be any antigenic determinant. [0061]
  • Prior to combination with the immunolabeling complexes, the sample is prepared in a way that makes the target materials in the sample accessible to the immunolabeling complexes. Typically, the samples used in the invention are comprised of tissue, cells, cell extracts, cell homogenates, purified or reconstituted proteins, recombinant proteins, biological fluids, or synthesized proteins. Large macromolecules such as immunolabeling complexes tend to be impermeant to membranes of live biological cells. Treatments that permeabilize the plasma membrane, such as electroporation, shock treatments, or high extracellular ATP, can be used to introduce the labeling mixture and its immunolabeling complexes into cells. Alternatively, the labeling mixture and its immunolabeling complexes can be physically inserted into cells, e.g. by pressure microinjection, scrape loading, patch-clamp methods, or phagocytosis. However, the target materials may require purification or separation prior to addition of the immunolabelling complexes, which will depend on the way the antigenic determinants are contained in the sample. When the sample contains purified target materials, the purified target materials may still be mixtures of different materials. For example, purified protein or nucleic acid mixtures may contain several different proteins or nucleic acids. Alternatively, the purified target materials may be electrophoresed on gels such as agarose or polyacrylamide gels to provide individual species of target materials that may be subsequently blotted onto a polymeric membrane or detected within the gel matrix. Preparation of a sample containing purified nucleic acids or proteins generally includes denaturation and neutralization. DNA may be denatured by incubation with base (such as sodium hydroxide) or heat. RNA is also denatured by heating (for dot blots) or by electrophoresing in the presence of denaturants such as urea, glyoxal, or formaldehyde, rather than through exposure to base (for Northern blots). Proteins are denatured by heating in combination with incubation or electrophoresis in the presence of detergents such as sodium dodecyl sulfate. The nucleic acids are then neutralized by the addition of an acid (e.g., hydrochloric acid), chilling, or addition of buffer (e.g., Tris, phosphate or citrate buffer), as appropriate. [0062]
  • Preferably, the preparation of a sample containing purified target materials further comprises immobilization of the target materials on a solid or semi-solid support. Purified nucleic acids are generally spotted onto filter membranes such as nitrocellulose filters or nylon membranes in the presence of appropriate salts (such as sodium chloride or ammonium acetate) for DNA spot blots. Alternatively, the purified nucleic acids are transferred to nitrocellulose filters by capillary blotting or electroblotting under appropriate buffer conditions (for Northern or Southern blots). To permanently bind nucleic acids to the filter membranes, standard cross-linking techniques are used (for example, nitrocellulose filters are baked at 80° C. in vacuum; nylon membranes are subjected to illumination with 360 nm light). The filter membranes are then incubated with solutions designed to prevent nonspecific binding of the nucleic acid probe (such as BSA, casein hydrolysate, single-stranded nucleic acids from a species not related to the probe, etc.) and hybridized to probes in a similar solution. Purified proteins are generally spotted onto nitrocellulose or nylon filter membranes after heat and/or detergent denaturation. Alternatively, the purified proteins are transferred to filter membranes by capillary blotting or electroblotting under appropriate buffer conditions (for Western blots). Nonspecifically bound probe is washed from the filters with a solution such as saline-citrate or phosphate buffer. Filters are again blocked, to prevent nonspecific adherence of immunolabeling complexes. Finally, samples are mixed with immunolabeling complexes. Nonspecifically bound immunolabeling complexes are typically removed by washing. [0063]
  • When the sample contains cellular nucleic acids (such as chromosomal or plasmid-borne genes within cells, RNA or DNA viruses or mycoplasma infecting cells, or intracellular RNA) or proteins, preparation of the sample involves lysing or permeabilizing the cell, in addition to the denaturation and neutralization already described. Cells are lysed by exposure to agents such as detergent (for example sodium dodecyl sulfate, Tween, sarkosyl, or Triton), lysozyme, base (for example sodium, lithium, or potassium hydroxide), chloroform, or heat. Cells are permeabilized by conventional methods, such as by formaldehyde in buffer. [0064]
  • As with samples containing purified target materials, preparation of the sample containing cellular target materials typically further comprises immobilization of the target materials on a surface such as a solid or semi-solid support. The targets may be arrayed on the support in a regular pattern or randomly. These supports include such materials as slides, beads, optical fibers, and membranes. The beads are preferably fluorescent or nonfluorescent polystyrene, the slides and optical fibers are preferably glass or plastic, and the membrane is preferably poly(vinylidene difluoride) or nitrocellulose. Thus, for example, when the sample contains lysed cells, cells in suspension are spotted onto or filtered through nitrocellulose or nylon membranes, or colonies of cells are grown directly on membranes that are in contact with appropriate growth media, and the cellular components, such as proteins and nucleic acids, are permanently bound to filters as described above. Permeabilized cells are typically fixed on microscope slides with known techniques used for in situ hybridization and hybridization to chromosome “squashes” and “spreads,” (e.g., with a reagent such as formaldehyde in a buffered solution). Alternatively, the samples used may be in a gel or solution. [0065]
  • Following the combining of the sample with the labeling mixture, unbound immunolabeling complexes that do not bind to the target are optionally removed from the sample by conventional methods, such as washing. The bound immunolabeling complex that binds to the target can be fixed in place with the usual fixatives (e.g. formaldehyde, glutaraldehyde) and fixation methods. Fixation can be utilized to improve the durability of the sample and to prevent transfer of the noncovalently complexed labeling protein to other targeting antibodies in the sample that have the same specific binding region. [0066]
  • Detection of the bound label is performed using methods and reagents well known to those skilled in the art. A preferred method of detection of the invention is through the use of fluorescence. Fluorescence from the immunolabeling complex binding to the target can be visualized with a variety of imaging techniques, including ordinary light or fluorescence microscopy, confocal laser-scanning microscopy, and flow cytometry, optionally using image deconvolution algorithms. Three-dimensional imaging resolution techniques in confocal microscopy utilize knowledge of the microscope's point spread function (image of a point source) to place out-of-focus light in its proper perspective. Multiple-labeled target materials are optionally resolved spatially, chronologically, by size, or using detectably different spectral characteristics (including excitation and emission maxima, fluorescence intensity, fluorescence lifetime, fluorescence polarization, fluorescence photobleaching rates, or combinations thereof), or by combinations of these attributes. Typically, multiple-labeled target materials are resolved using different labeling proteins with distinct spectral characteristics for each target material. Alternatively, the labels on the labeling proteins are the same but the samples are labeled and viewed sequentially or are spatially separated. [0067]
  • Additionally, enzymes can be used where there occurs an enzyme-dependent secondary generation of signal, such as the formation of a colored product from a non-colored substrate. Enzyme labels or enzyme labeling systems are desirable in that they can achieve signal amplification and greater distinctions from backgrounds. The enzyme breaks down a substrate to produce a chromophore or fluorophore or other detectable signal, thus amplifying the sensitivity of the assay and, if the substrate yields a distinct product at or near its site of formation, visualizing the site of the antigen/antibody complex in the sample. The substrate is selected to yield the preferred measurable product. Chromogenic, fluorogenic and chemiluminescence-generating enzyme substrates are preferred. These enzymes are enzymes for which substrates yielding useful chromophores, fluorophores, or chemiluminescence are known. Such substrates are extensively used in the art and are described the MOLECULAR PROBES HANDBOOK OF FLUORESCENT PROBES AND RESEARCH CHEMICALS by R. P. [0068] Haugland 6th Ed., (1996) and its subsequent 7th edition and 8th edition updates issued on CD Rom in November 1999 and May 2001, respectively, the contents of which are incorporated by reference, and in other published sources.
  • Preferred enzyme substrates of the invention are enzyme substrates that yield a fluorescent product that localizes at or near the site of enzyme activity. Enzymes of use in the method include any enzymes that utilize a chromogenic, fluorogenic, or chemiluminescence-generating substrate. Preferred enzymes of the invention include peroxidases, phosphatases, glycosidases, aequorins, or luciferases, and more specifically, HRP, [0069] Coprinus cinereus peroxidase, Arthromyces ramosus peroxidase, alkaline phosphatase, β-galactosidase, β-glucuronidase, or a protein A or protein G fusion protein of luciferase.
  • A preferred chromogenic (and in some cases fluorogenic) substrate and enzyme combination uses oxidoreductases such as horseradish peroxidase, [0070] Coprinus cinereus peroxidase, or Arthromyces ramosus peroxidase and a substrate such as 3,3′-diaminobenzidine (DAB) or 3-amino-9-ethylcarbazole (AEC), which yield a distinguishing color (brown and red, respectively). Other chromogenic oxidoreductase substrates that yield detectable products include, but are not limited to: 2,2′-azino-bis(3-ethylbenzthiazoline-6-sulfonic acid (ABTS), o-phenylenediamine (OPD), 3,3′,5,5′-tetramethylbenzidine (TMB), o-dianisidine, 5-aminosalicylic acid, and 4-chloro-1-naphthol. Fluorogenic substrates include, but are not limited to, homovanillic acid or 4-hydroxy-3-methoxyphenylacetic acid, reduced phenoxazines and reduced benzothiazines, including the Amplex Red reagent and its variants (Miike, U.S. Pat. No. 4,384,042), reduced dihydroxanthenes, including the Amplex Gold reagent and other dihydrofluoresceins such as those described in U.S. Pat. No. 6,162,931, and dihydrorhodamines such as dihydrorhodamine 123. Peroxidase substrates that are tyramides, as described in U.S. Pat. Nos. 5,196,306; 5,583,001 and 5,731,158, which are incorporated by reference, represent a unique class of peroxidase substrates in that they can be intrinsically detectable before action of the enzyme but are “fixed in place” by the action of a peroxidase in the process termed tyramide signal amplification (TSA). These substrates, which are a preferred embodiment of the instant invention, are extensively utilized to label targets in samples that are cells, tissues, or arrays for their subsequent detection by microscopy, flow cytometry, optical scanning, and fluorometry.
  • Another preferred chromogenic (and in some cases fluorogenic) substrate and enzyme combination uses a phosphatase enzyme such as calf intestinal alkaline phosphatase, an acid phosphatase, or a recombinant version of such a phosphatase in combination with a chromogenic substrate such as 5-bromo-4-chloro-3-indolyl phosphate (BCIP), 6-chloro-3-indolyl phosphate, 5-bromo-6-chloro-3-indolyl phosphate, p-nitrophenyl phosphate, or o-nitrophenyl phosphate or with a fluorogenic substrate such as 4-methylumbelliferyl phosphate, carboxyumbelliferyl phosphate, 6,8-difluoro-7-hydroxy4-methylcoumarinyl phosphate (DiFMUP, U.S. Pat. No. 5,830,912), fluorescein diphosphate, 3-O-methylfluorescein phosphate, resorufin phosphate, 9H-(1,3-dichloro-9,9-dimethylacridin-2-one-7-yl) phosphate (DDAO phosphate), or ELF 97, ELF 39, or related phosphates (U.S. Pat. Nos. 5,316,906 and 5,443,986). [0071]
  • Glycosidases, in particularly β-galactosidase, β-glucuronidase, and β-glucosidase, are additional suitable enzymes. Appropriate chromogenic substrates include, but are not limited to, 5-bromo-4-chloro-3-indolyl β-D-galactopyranoside (X-gal) and similar indolyl galactosides, glucosides, and glucuronides, o-nitrophenyl β-D-galactopyranoside (ONPG), and p-nitrophenyl β-D-galactopyranoside. Preferred fluorogenic glycosidase substrates include resorufm β-D-galactopyranoside, fluorescein digalactoside (FDG), fluorescein diglucuronide and their structural variants (U.S. Pat. Nos. 5,208,148; 5,242,805; 5,362,628; 5,576,424 and 5,773,236), 4-methylumbelliferyl β-D-galactopyranoside, carboxyumbelliferyl β-D-galactopyranoside, and fluorinated coumarin β-D-galactopyranosides (U.S. Pat. No. 5,830,912). [0072]
  • Additional enzymes include, but are not limited to, hydrolases such as cholinesterases and peptidases, oxidases such as glucose oxidase and cytochrome oxidases, and reductases, for which suitable substrates are known. [0073]
  • Enzymes and their appropriate substrates that produce chemiluminescence are preferred for some assays. These include, but are not limited to, natural and recombinant forms of luciferases and aequorins and include their chimeras with proteins that include protein A, protein G, and protein L. Chemiluminescence-producing substrates for phosphatases, glycosidases, and oxidases such as those containing stable dioxetanes, luminol, isoluminol, and acridinium esters are additionally useful. [0074]
  • When using florescent dyes to detect the desired target the sample is illuminated at a suitable absorption wavelength. A suitable wavelength is one that comes within the range of absorption wavelengths for each of the fluorescent dyes being used. Typically, the mixture is illuminated by a light source capable of producing light at or near the wavelength of maximum absorption of the dye or dyes, such as by ultraviolet or visible lamp, an arc lamp, a laser, or even sunlight. Illumination of the sample at a suitable wavelength results in one or more illuminated targets that are then analyzed according to the response of their fluorescence to the illumination. The illuminated targets are observed with any of a number of means for detecting a fluorescent response emitted from the illuminated target, including but not limited to visual inspection, cameras and film or other imaging equipment, or use of instrumentation such as fluorometers, plate readers, laser-based scanners, microscopes, or flow cytometers, or by means for amplifying the signal such as a photomultiplier (PMT). [0075]
  • In a particular aspect of the detection method, the labeling mixture is combined with a sample of cells in a fluid, such as ascites, hybridoma supernatant, or serum, and the presence or absence of the target in such cells is detected by using an automated instrument that sorts cells according to the detectable fluorescence response of the detectable moieties in the immunolabeling complexes bound to such cells, such as by fluorescence activated cell sorting (FACS), which is described in Mansour, et al., U.S. Pat. No. 4,665,024 (1987) and is incorporated by reference. In another aspect of the invention, additional detection reagents are combined with the sample concurrently with or following the labeling mixture. Such additional detection reagents include, but are not limited to reagents that selectively detect cells or subcellular components, ions, or indicate the cell viability, life cycle, or proliferation state. For example, the additional detection reagent is a labeled antibody that is directly or indirectly detectable and another additional detection reagent is a stain for nucleic acids, for F-actin, or for a cellular organelle. [0076]
  • While there are many applications in the field of immunological monitoring in which the presence of body fluid antibodies and antigens are detected by a variety of methods, these methods usually measure one antibody or antigen at a time and tend to be time consuming and costly. One of the strengths of the methods of the instant invention is the ability to simultaneously, but discretely, analyze multiple targets, especially wherein multiple antibodies from the same species (e.g. mouse monoclonal antibodies) are utilized. Another strength is the ability to rapidly prepare and optimize the complexes and to mix and combine colors of products for detection of the multiple targets in a sample. Determination of more than one target in a sample is accomplished with a labeling mixture containing more than one immunolabeling complex wherein a first target-binding antibody is bound to a labeling proteins with a first label and a second target-binding protein is bound to a labeling protein with a second label, wherein the first and second labels are detectably distinct. Thus multiple, distinct targets, can be determined simultaneously. Alternatively, any combination of labels, labeling proteins and target-binding antibodies may be used allowing for unlimited options. [0077]
  • In accordance with the subject invention compositions of the present invention include isolated immunolabeling complexes and isolated monovalent labeling proteins. The labeling protein is labeled with one or more labels. Typically, the labels that are attached to the labeling protein are directly detectable moieties, which directly detectable moieties are optionally the same or different, or the label is an enzyme or a hapten that is indirectly detectable. In a preferred embodiment of the invention the labeling protein is prepared by the process of (a) complexation of the unlabeled labeling protein with an immobilized form of its complementary binding protein; and (b) chemical conjugation of the labeling protein to the label; and (c) disaggregation of the covalently labeled labeling protein from the immobilized complementary protein. [0078]
  • Covalent conjugates of the enzyme and the labeling protein of the invention that are suitable for preparation of labeling complexes are typically prepared by methods well known in the art (MOLECULAR PROBES HANDBOOK OF FLUORESCENT PROBES AND RESEARCH CHEMICALS, Chapter 5 (1996)). Additionally, fusion proteins of enzymes such as of alkaline phosphatase, horseradish peroxidase, luciferase, and aequorin with the labeling proteins, protein A and protein G, have been described (Sun et al., J. Immunol. Meth. 152, 43 (1992); Eliasson et al., J. Biol. Chem. 263, 4323 (1988); Eliasson et al., J. Immunol. 142, 575 (1989)); U.S. Pat. No. 5,766,941 and 5,798,441; U.S. Pat. No. 5,292,658 and 5,418,155) that can be directly complexed with the target-binding antibody. Fusion proteins of green-fluorescent protein (GFP) and its wavelength-shifted analogs with certain immunoglobulin-binding proteins provide labeling proteins with a well-defined stoichiometry and structure. Labeling proteins that are conjugates of phycobiliproteins (e.g. U.S. Pat. Nos. 4,520,110; 4,859,582; 5,055,556; all incorporated by reference) or of tandem conjugates of phycobiliproteins with additional dyes (e.g. U.S. Pat. No. 4, 542,104; incorporated by reference) are prepared by similar coupling methods. [0079]
  • Chemical labeling of the monovalent antibody fragment is optionally performed while the antibody fragment is immobilized on a matrix, such as while bound to its complementary binding site. For example, an Fab fragment of goat anti-(mouse Fc) or a protein A or protein G can be bound to a whole mouse IgG that has been immobilized on agarose. Following reaction with a reactive dye, or coupling to a protein such as a phycobiliprotein, a tandem conjugate of a phycobiliprotein with an Alexa Fluor dye (Molecular Probes, Inc.) or with an enzyme by standard methods, the excess reactive dye is removed while the monovalent labeling protein is still immobilized and then the labeled monovalent antibody is eluted by reducing the pH. This method has the simultaneous advantages of facilitating elimination of the unconjugated reactive dye and protecting the combining region of the monovalent antibody. Furthermore, we have observed that fluorescent dye conjugates prepared by this indirect method typically have greater fluorescence or activity than do conjugates of the monovalent labeling protein prepared using the same reactive dye in a homogeneous solution. We have also observed that less labeled monovalent labeling protein is required to achieve the same or superior detectability when preparing a labeling complex with the target-binding antibody. Alternatively, smaller quantities of target-binding antibody can be used when pre-forming the immunolabeling complexes to achieve the same detectablity. Optionally, the target-binding antibody is about 5 micrograms to about 0.001 micrograms. [0080]
  • The invention also includes an isolated protein G complex consisting essentially of protein G and a non-covalently bound albumin. The albumin is covalently labeled with one or more detectable labels, as described above, where the detectable labels are optionally the same or different. Various types of albumin can be used including, for example, human albumin, bovine serum albumin, or ovalbumin. [0081]
  • In yet another aspect of the invention, the isolated immunolabeling complex is made by combining, in vitro, one or more target-binding antibodies with an excess of labeling protein that has been covalently labeled as described above, under conditions suitable for the labeling protein to bind selectively and with high affinity to a selected region of said target-binding antibody to form an immunolabeling complex, and isolating said immunolabeling complex. In one example of forming the complex, combining means mixing a solution of the isolated covalently labeled labeling protein with a solution of the target-binding antibody in a suitable buffer for a period sufficient to form a noncovalent complex. Typically this period is under five minutes but may be less than one minute. [0082]
  • When preparing the immunolabeling complex, stock solutions of both the labeling protein and the target-binding antibody are typically near 1 mg/mL in an appropriate buffer, although more or less concentrated solutions are also suitable. Generally, the labeling protein is mixed in a molar ratio of at least one to 50 moles of labeling protein to one mole of the region of the target-binding antibody to be complexed. More commonly a ratio of at least one to as many as 10 moles of labeling protein per mole of target-binding antibody is combined. With an Fab fragment of an antibody to the Fc region of a target-binding antibody, a molar ratio of approximately 2 to 10 is typical, more typically 3 to 5 (particularly for complexes in which the labeling protein has been labeled while immobilized on an affinity matrix). The ease of formation of the complex permits rapid optimization of the complex and assessment of the effect of variation in experimental parameters. A particularly unique advantage of the invention is that the stoichiometry of the complex is easily adjusted to provide complexes with different ratios of labeling protein to target-binding antibody, and thus there is some control over the ultimate detectability of the target in the sample. This feature is impractical with primary antibodies that have been directly chemically labeled with dyes. Complexes that have been labeled with the same dye but at different molar ratios can be separately detected by the differences in their intensities. [0083]
  • Complex formation appears to occur almost within the mixing time of the solutions (<1 minute) but the reaction typically is allowed to proceed for at least 5 minutes and can be longer before combining the antibody complex with the sample. Although complex formation can be reversed by addition of an unlabeled antibody that contains the same binding region, reversibility is very slow; furthermore, following staining of the sample with the complex, the sample can be “fixed” using aldehyde-based fixatives by methods that are commonly practiced by those skilled in the art of immunolabeling. [0084]
  • A unique and important advantage of the method of the invention is the ability to form the immunolabeling complex from exceedingly small quantities of the target-binding antibody and labeling protein, which is impractical when using chemically reactive dyes. Complexes of the invention have been prepared from sub-micrograms of a target-binding antibody with sub-micrograms of a labeling protein. The process of forming a labeling complex can easily be automated. An important use of this characteristic is that of screening hybridoma supernatants in the generation and optimization of new antibodies, where the very small quantity of antibody present in a single well makes it impractical to perform chemical labeling. [0085]
  • In one aspect of the invention, a capture component is added to the labeling mixture, prior to combining the labeling mixture with the sample, to remove excess labeling protein. For applications in which immunolabeling complexes of multiple primary antibodies from the same species (e.g. mouse monoclonal antibodies) or cross-reacting species (e.g. mouse and human antibodies) are to be used simultaneously or sequentially, it is necessary to quench or otherwise remove any excess labeling protein by use of a capture component or by other means to avoid inappropriate labeling of the sample. The most effective capturing components to capture excess labeling protein are those that contain the binding site of the labeling protein but are themselves not labeled, preferably an antibody or antibody fragment. Capture components may be free in solution or immobilized on a solid phase, such as agarose, cellulose, or a natural or synthetic polymer, to facilitate separation of the excess capture component from the labeled protein. The capture component is optionally attached to a microsphere or magnetic particle. Any of the isolated labeling mixtures optionally include a capture component to remove excess labeling protein. However, separation of excess labeling protein is not essential for successful utilization of the invention, particularly when using a single target-binding antibody. [0086]
  • Included in the invention are isolated labeling mixtures comprising one or more immunolabeling complexes, wherein at least one immunolabeling complex is comprised of a target-binding antibody, is selective for a target in a sample, and a labeling protein, as described above, that binds selectively and with high affinity to a selected region of the target-binding antibody. More preferably the labeling mixture contains at least three such complexes. The potential number of immunolabeling complexes that can be used simultaneously is principally limited by the number of targets for which selective antibodies are available and the ability to spatially or spectrally resolve the detectable signals, such as on a Western blot, a protein array, a chromosome “spread”, or a microsphere or “chip” array (e.g., U.S. Pat. Nos. 5,981,180 and 5,736,330). [0087]
  • These labeling mixtures optionally include additional detection reagents such as those known to those skilled in the art, including reagents that localize the same or additional targets in the sample. Preferred additional detection reagents include stains for cell organelles, reagents to indicate the state of cell viability or proliferation, and enzyme substrates. Other preferred additional detection reagents are additional antibodies that can be detected in conjunction with direct or indirect labeling methods, wherein the targets of such additional antibodies and of the target-binding antibody are optionally the same or different. [0088]
  • In one aspect of the invention, the labeling protein is a monovalent fragment that is made by (a) generating a labeling antibody against an antigen, where said antigen is a Fc region of one or more isotypes of an antibody from a different species; (b) digesting said labeling antibody to generate multiple monovalent fragments; (c) covalently labeling said monovalent fragments with one or more labels; and (d) isolating the labeled fragments. [0089]
  • Particularly preferred for use with mouse or rat monoclonal antibodies is an Fab fragment of an antibody to mouse Fc, wherein the whole antibody was produced in goat, chicken or other species. Preferred as a labeling protein for complexing with either targeting or secondary antibodies produced in other species (e.g. rabbit, sheep, goat, donkey, bovine, horse, guinea pig, chicken) are antibodies to the Fc region of these target-binding or secondary antibodies produced in a non-cross-reacting animal, in particular Fab fragments of such antibodies. Particularly where the primary antibody to be labeled is a mouse, rat, or human antibody, additional selectivity can be obtained by utilizing an antibody directed at a particular isotype of the primary antibody, such as toward IgG[0090] 1, IgG2a, or IgG3 isotypes.
  • The target-binding antibody is selected based on the desired target, where the target is an antigen selected for investigation or analysis. The chemical identity of the target antigen may be known or unknown. The target is optionally a material of biological or synthetic origin that is present as a molecule or as a group of molecules. Typically, the target is a biological material or antigenic determinant. Biological materials include, but are not limited to, antibodies, amino acids, proteins, peptides, polypeptides, enzymes, enzyme substrates, hormones, lymphokines, metabolites, antigens, haptens, lectins, avidin, streptavidin, toxins, poisons, environmental pollutants, carbohydrates, oligosaccharides, polysaccharides, glycoproteins, glycolipids, nucleotides, oligonucleotides, nucleic acids and derivatized nucleic acids (including deoxyribo- and ribonucleic acids and peptide nucleic acids), DNA and RNA fragments and derivatized fragments (including single and multi-stranded fragments), natural and synthetic drugs, receptors, virus particles, bacterial particles, virus components, biological cells, cellular components (including cellular membranes and organelles), natural and synthetic lipid vesicles, and polymer membranes. Typically the target material is present as a component or contaminant of a sample taken from a biological or environmental system. [0091]
  • III. Kits [0092]
  • Suitable kits for preparing a labeling protein, for preparing an immunolabeling complex, and for immunolabeling a target in a sample also form part of the invention. Such kits can be prepared from readily available materials and reagents and can come in a variety of embodiments. The contents of the kit will depend on the design of the assay protocol or reagent for detection or measurement. Generally, the kits will contain instructions, appropriate reagents and labels, and solid supports, as needed. Typically, instructions include a tangible expression describing the reagent concentration or at least one assay method parameter such as the relative amounts of reagent and sample to be admixed, maintenance time periods for reagent/sample admixtures, temperature, buffer conditions and the like to allow the user to carry out any one of the methods or preparations described above. Where a kit is designed for a simple detection assay for a target using a single highly specific antibody, the kit contains this highly specific antibody pre-complexed with a labeling protein. Alternatively, the kit comprises panels of antibodies each pre-complexed with a unique labeling protein whose recognition pattern for the desired target is calibrated or the kit comprises panels of antibodies and a separate labeling protein or labeling proteins. A labeled competitive mimotope or mixture is optionally additionally included to permit the assays to be conducted by competition of the unlabeled target in the sample with the labeled mimotope. [0093]
  • A kit for preparation of optimal labeling proteins typically comprises (a) a monovalent antibody fragment that binds to a specific region of a target-binding antibody, (b) a chemically reactive dye, and (c) an immobilization matrix comprising the specific region of the target-binding antibody to which the monovalent antibody fragment binds. [0094]
  • In one embodiment of the invention, the kit comprises (a) a mixture of one or more monovalent antibody fragments that bind to a specific region of one or more antibody isotypes, where each fragment is covalently labeled with one or more labels, (b) a capture component to which the antibody fragments bind, and optionally (c) comprises one or more target-binding antibodies. Preferably the capture component is purified mouse IgG or serum from a non-immune mouse and the label is a fluorescent molecule. In another preferred embodiment of this kit, the antibody fragment is an antibody to the Fc region of the target-binding antibody that is matched to or cross-reacts with the species from which the target-binding antibody is derived. In a further preferred embodiment of the kit, the labeled monovalent antibody fragment has been labeled while immobilized on an affinity matrix. [0095]
  • In another embodiment, a kit is provided for preparing an immunolabeling complex comprising another version of the kit for preparing (a) an immunolabeling complex that comprises a protein G complexed with a labeled albumin and (b) a capture component to which the protein G complexed with albumin binds. In a more particular embodiment of this kit, the capture component is purified mouse IgG or non-immune mouse serum and the albumin is human albumin, bovine serum albumin, or ovalbumin. In a more preferred embodiment the albumin is ovalbumin. [0096]
  • Another kit of the invention that is useful for immunolabeling a target in a sample comprises a labeling mixture that contains one or more of the immunolabeling complexes as defined above and a buffer. In a more preferred embodiment, the kit contains three or more such immunolabeling complexes. [0097]
  • Yet another kit of the invention that is useful for immunolabeling a target in a sample comprises a labeling mixture that contains one or more of the immunolabeling complexes, as defined above, a buffer, and additional labeling reagents that optionally include (a) directly conjugated target-binding antibodies that may be the same or different, (b) stains for characterization of cellular organelles, cell viability, or cell proliferation state, or (c) enzyme substrates. [0098]
  • A wide variety of kits and components can be prepared according to the present invention, depending upon the intended user of the kit and the particular needs of the user. [0099]
  • IV. Applications [0100]
  • The instant invention has useful applications in basic research, high-throughput screening, immunohistochemistry, fluorescence in situ hybridization (FISH), microarray technology, diagnostics, and medical therapeutics. The invention can be used in a variety of assay formats for diagnostic applications in the disciplines of microbiology, immunology, hematology and blood transfusion, tissue pathology, forensic pathology, and veterinary pathology. The invention is particularly useful in the characterization and selection of optimized antibodies from hybridoma supernatants. Additionally, the invention can be used to deliver therapeutics to a specific target. In general, the current invention provides a versatile and convenient method to enhance any assay that uses an antibody as part of its detection methodology. [0101]
  • The instant invention can be used to study biological phenomena, such as, for example, cell proliferation, signal transduction in cells, or apoptosis. For illustration purposes only and not limitation, one could study thymidine analog 5-bromo-2′-deoxyuridine (BrdU) incorporation. BrdU is a marker for both cell proliferation and apoptosis, as it is readily incorporated into newly synthesized DNA that has progressed through the S-phase of the cell cycle and also into DNA break sites by deoxynucleotidyl transferase (TdT). Anti-BrdU antibodies are used to detect cells marked by BrdU incorporation. By being able to directly label the anti-BrdU antibodies, the current invention provides a convenient method to allow for detection of the incorporated BrdU by conventional immunohistochemistry or fluorescence, depending on detection method required. [0102]
  • Additionally, the current invention has the advantage of allowing staining for multiple targets in one cocktail, thereby reducing the need for more samples or processing steps per experiment. This is particularly important when analyzing precious samples (e.g., pediatric samples, leukocytes isolated from biopsies, rare antigen-specific lymphocytes and mouse tissues that yield a small number of cells). Although it is currently possible to simultaneously measure up to 11 distinct fluorescent colors through a convoluted series of novel developments in flow cytometry hardware, software, and dye chemistry, the use of these advances has been severely limited by the lack of commercial availability of spectrally distinct directly labeled primary and secondary antibodies. Although labeled secondary antibodies directed at individual isotype-specific targeting antibodies (e.g., anti-IgG[0103] 1 isotype antibodies) exist, it is not possible to use this type of labeled antibody to detect more than one of the same isotype of an antibody (e.g., an IgG1 isotype antibody) in a single sample due to cross-reactivity. The current invention overcomes these limitations by providing for a convenient and extremely versatile method of rapidly labeling either small or large quantities of any primary antibody including primary antibodies of the same isotype to be used in, for example, multicolor flow cytometry and on Western blots. This advance in multicolor systems has a number of advantages over current two- and three-color flow cytometric measurements. For example, no combination of one-color stains can accurately enumerate or be used to isolate CD3+ CD4+ CD8 T cells (excluding, for example CD3+ CD4+ CD8+ T cells and small CD4+ monocytes). The use of cell membrane markers to study leukocyte composition in blood and tissue serves as an example of an analytical monoclonal antibody application, particularly in combination with flow cytometry. It is also the example most relevant to studies of the immune system, because the cellular composition of blood and lymphoid tissue provides a ‘window’, allowing the analysis and monitoring of the immune system.
  • The methods of the invention can also be used in immunofluorescence histochemistry. Immunofluorescence histochemistry involves the use of antibodies labeled with fluorophores to detect substances within a specimen. The pathologist derives a great deal of information of diagnostic value by examining thin sections of tissue in the microscope. Tissue pathology is particularly relevant to, for example, the early diagnosis of cancer or premalignant states, and to the assessment of immunologically mediated disorders, including inflammation and transplant rejection. The problems associated with immunofluorescence histochemistry, however, stem from the limitations of the methods currently available for use in such application. For example, directly labeling an antibody can result in antibody inactivation and requires a relatively large of amount of antibody and time to do the conjugation. It is also expensive and impractical to prepare directly labeled antibodies having variable degrees of label substitution. Similarly, indirect labeling of an antibody has problems, such as lack of secondary antibody specificity, and reliance upon primary antibody differences, including antibody isotypes and available fluorophores, to do multicolor labeling. Secondary antibody labeling is not practical where the primary antibody is from the same species or of the same isotypes. Combinations of fluorophores or other detectable labels on the same target-binding antibody, which can be readily prepared in multiple mixtures by the methods on this invention, greatly increase the number of distinguishable signals in multicolor protocols. Lack of secondary antibody specificity arises when the specimen containing the targeted moiety and target-binding antibody are from homologous species. For example, BrdU-labeled DNA in rodent tissue is detected by immunohistochemical staining. The target-binding antibody is conventionally mouse anti-BrdU, and the detecting antibody system uses an anti-mouse immunoglobulin antibody, labeled with fluorescein. Because there is homology between mouse immunoglobulin and immunoglobulins from a number of rodent species (for example, rats, mice, hamsters, etc.), the detecting antibody not only binds to the target-binding antibody, but also nonspecifically binds to immunoglobulin in the tissue. The current invention eliminates this problem by pre-forming the immunolabeling complex and allows for a simple, rapid and convenient method to proceed with labeling with two, three or more fluorescent antibodies in one experiment. Very significantly, it can always be used with primary antibodies of either the same or different isotype, and always on tissue of the same or similar species as the primary antibody. [0104]
  • The instant invention also has application in the field of microarrays. Microarray technology is a powerful platform for biological exploration (Schena (Ed.), Microarray Biochip Technology, (2000)). Many current applications of arrays, also known as “biochips,” can be used in functional genomics as scientists seek characteristic patterns of gene expression in different physiopathological states or tissues. A common method used in gene and protein microarray technology involves the use of biotin, digoxigenin (DIG), or dinitrophenyl (DNP) as an epitope or a “tag” such as an oligohistidine, glutathione transferase, hemagglutinin (HA), or c-myc. In this case a detectably labeled anti-biotin, anti-DIG, anti-DNP, anti-oligohistidine, anti-glutathione transferase, anti-HA, or anti-c-myc is used as the detection reagent. The instant invention allows for the use of multiple fluorophore- or enzyme-labeled antibodies, thereby greatly expanding the detection modalities and also providing for enhanced multiplexing and two-dimensional analysis capabilities. [0105]
  • Similarly, the invention can be used with protein microarrays and on Western blots. Protein microarrays can provide a practical means to characterize patterns of variation in hundreds of thousands of different proteins in clinical or research applications. Antibody arrays have been successfully employed that used a set of 115 antibody/antigen pairs for detection and quantitation of multiple proteins in complex mixtures (Haab et al., Genome Biology, 2, 4.1 (2001)). However, protein microarrays use very low sample volumes, which historically have significantly limited the use of antibody technology for this application. The invention of the application readily overcomes this limitation and provides a means to label antibodies with the fluorescent dyes using a very low sample volume and to automate formation of the staining complex and the staining process. [0106]
  • The present invention also provides a means for the specific detection, monitoring, and/or treatment of disease and contemplates the use of immunolabeling complexes to detect the presence of particular targets in vitro. In such immunoassays, the sample may be utilized in liquid phase, in a gel, or bound to a solid-phase carrier, such as an array of fluorophore-labeled microspheres (e.g., U.S. Pat. No. 5,981,180 and 5,736,330). For example, a sample can be attached to a polymer, such as aminodextran, in order to link the sample to an insoluble support such as a polymer-coated bead, plate, or tube. For instance, but not as a limitation, using the methods of the present invention in an in vitro assay, antibodies that specifically recognize an antigen of a particular disease are used to determine the presence and amounts of this antigen. [0107]
  • Likewise, the immunolabeling complexes of the present invention can be used to detect the presence of a particular target in tissue sections prepared from a histological specimen. Preferably, the tissue to be assayed will be obtained by surgical procedures, e.g., biopsy. The excised tissue will be assayed by procedures generally known in the art, e.g. immunohistochemistry, for the presence of a desired target that is recognized by an immunolabeling complex, as described above. The tissue may be fixed or frozen to permit histological sectioning. The immunolabeling complex may be labeled, for example with a dye or fluorescent label, chemical, heavy metal or radioactive marker to permit the detection and localization of the target-binding antibody in the assayed tissue. In situ detection can be accomplished by applying a detectable immunolabeling complex to the tissue sections. In situ detection can be used to determine the presence of a particular target and to determine the distribution of the target in the examined tissue: General techniques of in situ detection are well known to those of ordinary skill. See, for example, Ponder, “Cell Marking Techniques and Their Application,” in MAMMALIAN DEVELOPMENT: A PRACTICAL APPROACH, Monk (ed.), 115 (1987). [0108]
  • For diagnosing and classifying disease types, tissues are probed with an immunolabeling complex, as defined above, that comprises a target-binding antibody to a target antigen associated with the disease, e.g., by immunohistochemical methods. Where the disease antigen is present in body fluids, such immunolabeling complexes comprising a target-binding antibody to the disease antigen are preferably used in immunoassays to detect a secreted disease antigen target. [0109]
  • Detection can be by a variety of methods including, for example, but not limited to, flow cytometry and diagnostic imaging. When using flow cytometry for the detection method, the use of microspheres, beads, or other particles as solid supports for antigen-antibody reactions in order to detect antigens or antibodies in serum and other body fluids is particularly attractive. Flow cytometers have the capacity to detect particle size and light scattering differences and are highly sensitive fluorescence detectors. Microfluidic devices provide a means to perform flow-based analyses on very small samples. [0110]
  • Alternatively, one can use diagnostic imaging. The method of diagnostic imaging with radiolabeled antibodies is well known. See, for example, Srivastava (ed.), RADIOLABELED MONOCLONAL ANTIBODIES FOR IMAGING AND THERAPY, Plenum Press (1988); Chase, “Medical Applications of Radioisotopes,” in REMINGTON'S PHARMACEUTICAL SCIENCES, 18th Edition, Gennaro et al. (eds.) Mack Publishing Co., 624 (1990); and Brown, “Clinical Use of Monoclonal Antibodies,” in BIOTECHNOLOGY AND PHARMACY, Pezzuto et al. (eds.), Chapman & Hall, 227 (1993). This technique, also known as immunoscintigraphy, uses a gamma camera to detect the location of gamma-emitting radioisotopes conjugated to antibodies. Diagnostic imaging is used, in particular, to diagnose cardiovascular disease and infectious disease. [0111]
  • Thus, the present invention contemplates the use of immunolabeling complexes to diagnose cardiovascular disease. For example, immunolabeling complexes comprising anti-myosin antibodies can be used for imaging myocardial necrosis associated with acute myocardial infarction. Immunolabeling complexes comprising antibodies that bind platelets and fibrin can be used for imaging deep-vein thrombosis. Moreover, immunolabeling complexes comprising antibodies that bind to activated platelets can be used for imaging atherosclerotic plaque and immunolabeling complexes. [0112]
  • Immunolabeling complexes of the present invention also can be used in the diagnosis of infectious diseases. For example, immunolabeling complexes comprising antibodies that bind specific bacterial antigens can be used to localize abscesses. In addition, immunolabeling complexes comprising antibodies that bind granulocytes and inflammatory leukocytes can be used to localize sites of bacterial infection. Similarly, the immunolabeling complexes of the present invention can be used to detect signal transduction in cells, the products of signal transduction, and defects, inhibitors, and activators of signal transduction. [0113]
  • Numerous studies have evaluated the use of antibodies for scintigraphic detection of cancer. Investigations have covered the major types of solid tumors such as melanoma, colorectal carcinoma, ovarian carcinoma, breast carcinoma, sarcoma, and lung carcinoma. Thus, the present invention contemplates the detection of cancer using immunolabeling complexes comprising antibodies that bind tumor markers (targets) to detect cancer. Examples of such tumor markers include carcinoembryonic antigen, α-fetoprotein, oncogene products, tumor-associated cell surface antigens, and necrosis-associated intracellular antigens. In addition to diagnosis, antibody imaging can be used to monitor therapeutic responses, detect recurrences of a disease, and guide subsequent clinical decisions and surgical procedures. In vivo diagnostic imaging using fluorescent complexes that absorb and emit light in the near infrared (such as those of the Alexa Fluor 700 and Alexa Fluor 750 dyes) is also known. [0114]
  • In addition to use in diagnostic applications, the invention contemplates the use of immunolabeling complexes as therapeutics. Immunolabeling complexes that are therapeutically substituted or therapeutic immunolabeling complexes, wherein the labeling protein is alternatively conjugated with a radioactive moiety, a toxin, or a drug, as opposed to with a detectable label, to form a drug-labeled protein or immunoconjugate, can be used to treat viral and bacterial infectious diseases, cardiovascular disease, autoimmune disease, and cancer. The objective of such therapy is to deliver cytotoxic doses of radioactivity, toxin, or drug to target cells, while minimizing exposure to non-target tissues. [0115]
  • Therapeutic agents useful in the treatment of a disease may be conjugated to the protein by methods known to those of skill in the art. Examples of such therapeutic agents include peptides; proteins; small organic drugs, such as, doxorubicin, daunorubicin, methotrexate, melphalin, chlorambucil, vinca alkaloids, 5-fluorouridine, and mitomycin-C; radioisotopes, such as, [0116] 131I, 67Cu, or 90Y; and cytotoxic agents, such as ricin, abrin, pokeweed antiviral protein, gelonin, diphtherin toxin, and Pseudomonas endotoxin. 131I can be covalently bound to the protein by methods known in the art, e.g., lodogen reactions (Frank et al., Biochem. Biophys. Res. Commun. 80, 849 (1978)). 67Cu, one of the more promising radioisotopes for radioimmunotherapy due to its 61.5 hour half-life and abundant supply of beta particles and gamma rays, can be conjugated to the protein using the chelating agent, 6-(p-bromoacetamidobenzyl)-1,4,8,1 1-tetraazacyclotetradecane-N,N′,N″,N′″-tetraacetic acid (TETA). Alternatively, 90Y, which emits an energetic beta particle, can be coupled to a labeling protein using diethylenetriaminepentaacetic acid (DTPA), or more preferably, 1,4,7,10-tetraazacyclododecane-N,N′,N″,N′″-tetraacetic acid (DOTA) (Camera et al., J. Nucl. Med. 35, 882 (1994)). Agents such as glutaraldehyde or a carbodimide may conjugate the toxins to the protein. Small organic drugs, peptides, and proteins can be conjugated by various means known in the art, for example, but not limitation, through amide bonds and or linkers.
  • Administration of the therapeutically substituted immunolabeling complex may be by various means known in the art, but generally will be by injection, intravenous, intraarterial, intraperitoneal, intramuscular, subcutaneous, intrapleural, intrathecal, by perfusion through a regional catheter, or by direct intralesional injection. When administering therapeutic immunolabeling complexes by injection, the administration may be by continuous infusion, or by single or multiple boluses. [0117]
  • The therapeutic immunolabeling complex of the present invention can be formulated according to known methods to prepare pharmaceutically useful compositions, whereby the therapeutic immunolabeling complexes are combined in a mixture with a pharmaceutically acceptable carrier. A composition is said to be a “pharmaceutically acceptable carrier” if its administration can be tolerated by a recipient patient. Sterile phosphate-buffered saline is one example of a pharmaceutically acceptable carrier. Other suitable carriers are well known to those in the art. See, for example, REMINGTON'S PHARMACEUTICAL SCIENCES, 18th Ed. (1990). [0118]
  • For purposes of immunotherapy, a substituted immunolabeling complex and a pharmaceutically acceptable carrier are administered to a patient in a therapeutically effective amount. A combination of an immunolabeling complex and a pharmaceutically acceptable carrier is said to be administered in a “therapeutically effective amount” if the amount administered is physiologically significant. An agent is physiologically significant if its presence results in a detectable change in the physiology of a recipient patient. A useful therapeutic dose will vary with the particular therapeutic agent used, the particular disease type and history, and the specific considerations of the patient to be treated, such as the patient's age, weight, height, sex, general medical condition, and previous medical history. A physician administering the agent will know to calculate the effective therapeutic dose, which dose will be effective in reducing or eliminating the disease without compromising significantly normal tissues or cells of the patient. Typically, however, it is desirable to provide the recipient with a dosage of the substituted immunolabeling complex that is in the range of from about 1 pg/kg to 10 mg/kg (amount of agent/body weight of patient), although a lower or higher dosage may also be administered. For example, many studies have demonstrated successful treatment with doses of 0.1 to 1.0 milligram, while other studies have shown improved localization with doses in excess of 10 milligrams. [0119]
  • Additional pharmaceutical methods may be employed to control the duration of action of a substituted immunolabeling complex in a therapeutic application. Controlled release preparations can be prepared through the use of polymers to complex or adsorb a substituted immunolabeling complex. For example, biocompatible polymers include matrices of poly(ethylene-co-vinyl acetate) and matrices of a polyanhydride copolymer of a stearic acid dimer and sebacic acid (Sherwood et al., [0120] Biotechnology 10, 1446 (1992). The rate of release of a substituted immunolabeling complex from such a matrix depends upon the molecular weight of the substituted immunolabeling complex, the amount of substituted immunolabeling complex within the matrix, and the size of dispersed particles (Saltzman et al., Biophysical J. 55, 163 (1989). Other solid dosage forms are described in REMINGTON'S PHARMACEUTICAL SCIENCES, 18th Ed. (1990). Each of the above-described references is incorporated herein by reference in its entirety.
  • EXAMPLES
  • The following examples describe specific aspects of the invention to illustrate the invention and to provide a description of the methods for those of skill in the art. The examples should not be construed as limiting the invention, as the examples merely provide specific methodology useful in understanding and practicing the invention. [0121]
  • Example 1 Preparation of Mouse Fc Antigen
  • Purified mouse IgG was fragmented with the proteolytic enzyme papain (CURRENT PROTOCOLS IN CELL BIOLOGY, 16.4.1-16.4.10 (2000)). A 12 mL solution of mouse IgG was prepared at ˜2 mg/mL in phosphate-buffered saline (PBS). A solution containing 0.1 mg of papain in digestion buffer (PBS, 0.02 M EDTA, 0.02 M cysteine) was added to the antibody and allowed to react at 37° C. for 16 hours. The digestion was terminated by the [0122] addition 20 μL of 0.3 M iodoacetamide in PBS. The fragments were dialyzed against 2 L of PBS for 16 hours at 4° C. The Fc fragment was purified on a protein G-Sepharose CL-4B column. The bound fraction containing the Fc fragment was eluted from the column using 50-100 mM glycine/HCl buffer, pH 2.5-2.8. The eluate was collected in 1 mL fractions. The pH of the protein fractions was immediately raised to neutral by addition of 100 μL of either 500 mM phosphate or Tris buffer, pH 7.6, to each 1 mL fraction. The solution was then loaded onto a Sephacryl S-200 Superfine size-exclusion column and fractions corresponding to a molecular weight of ˜50 kDa were collected and analyzed by SDS-PAGE and HPLC. The goat anti-(rabbit Fc) fragment was prepared similarly.
  • Example 2 Goat Anti-(Mouse Fc) Production
  • To produce polyclonal antibodies, the immunogen used to immunize the animal was the purified mouse or rabbit IgG Fc domain (see Example 1). Methods of immunizing animals are well known and conventional, and suitable immunization protocols and immunogen concentrations can be readily determined by those skilled in the art (Current Protocols in Immunology 2.4.1-9 (1995); ILAR Journal 37, 93 (1995)). Briefly, individual goats were immunized with purified mouse Fc or purified rabbit Fc. The initial immunization in 50% Freund's complete adjuvant (1000 μg conjugate (half subcutaneous, half intramuscularly)) was followed by 500 μg conjugate per goat in Freund's incomplete adjuvant two and four weeks later and at monthly intervals thereafter. Antibodies were harvested from serum using protein A-Sepharose chromatography. Antibodies against mouse Fc isotypes can be prepared by starting with isotype-selected mouse Fc antigens. Rabbits have a single Fc isotype. Characterization of the selectivity and cross-reactivity of isotype-specific antibodies is by standard techniques, including HPLC. [0123]
  • Example 3 Preparation of Fab Fragments
  • Fragmentation of the goat anti-(mouse Fc) antibody to the monovalent Fab fragment was carried out using the proteolytic enzyme, papain, as described in Example 1. Following dialysis against PBS, the Fab fragment was purified on a protein A-Sepharose CL-4B column. The unbound fraction containing the Fab fragment and the papain was collected. This solution was then loaded onto a Sephacryl S-200 Superfine size-exclusion column and fractions corresponding to a molecular weight of ˜50 kDa were collected and analyzed by SDS-PAGE. The Fab fragments of goat anti-(rabbit Fc) can be prepared similarly. [0124]
  • Example 4 Preparation of the Labeled Antibody Immunoglobulin-Binding Protein or the Non-Antibody Immunoglobulin-Binding Peptide and Protein Conjugates in Homogeneous Solution
  • Conjugates of antibody immunoglobulin-binding protein or the non-antibody immunoglobulin-binding peptides or proteins with low molecular weight dyes and haptens such as biotin or digoxigenin are typically prepared from succinimidyl esters of the dye or hapten, although reactive dyes and haptens having other protein-reactive functional groups are also suitable. The typical method for protein conjugation with succinimidyl esters is as follows. Variations in molar ratios of dye-to-protein, protein concentration, time, temperature, buffer composition and other variables that are well known in the art are possible that still yield useful conjugates. [0125]
  • A protein solution of the Fab fragment of goat anti-(rabbit Fc), goat anti-(mouse Fc), protein A, protein G, or protein L or an immunoglobulin-binding peptide (e.g., a peptide identified by screening a library of peptides) is prepared at ˜10 mg/mL in 0.1 M sodium bicarbonate (pH ˜8.3). The labeling reagents are dissolved in a suitable solvent such as DMF at ˜10 mg/mL. Predetermined amounts of the labeling reagents are added to the protein solution with stirring. A molar ratio of 10 moles of dye to 1 mole of protein is typical, though the optimal amount can be varied with the particular labeling reagent, the protein being labeled and the protein's concentration. The optimal ratio was determined empirically. When optimizing the fluorescence yield and determining the effect of degree of substitution (DOS) on the conjugate's brightness, it is typical to vary the ratio of reactive dye to protein over a several-fold range. The reaction mixture is incubated at room temperature for a period that is typically one hour or on ice for several hours. The dye-protein conjugate is typically separated from unreacted reagents by size-exclusion chromatography, such as on BIO-RAD P-30 resin equilibrated with PBS. The initial, protein-containing band is collected and the DOS is determined from the absorbance at the absorbance maximum of each fluorophore, using the extinction coefficient of the free fluorophore. The DOS of nonchromophoric labels, such as biotin, is determined as described in Haugland (Haugland et al., Meth. Mol. Biol. 45, 205 (1995); Haugland, Meth. Mol. Biol. 45, 223 (1995); Haugland, Meth. Mol. Biol. 45, 235 (1995); Haugland, Current Protocols in Cell Biol. 16.5.1-16.5.22 (2000)). Using the above procedures, conjugates of goat anti-(mouse Fc) and goat anti-(rabbit Fc) were prepared with several different Alexa Fluor dyes, with Oregon Green dyes, with biotin-X succinimidyl ester, with desthiobiotin-X succinimidyl ester, with succinimidyl 3-(2-pyridyldithio)propionate (SPDP) and with succinimidyl trans-4-(maleimidylmethyl)cyclohexane-1-carboxylate (SMCC). [0126]
  • Some dye conjugates of protein A and protein G, including those of some Alexa Fluor dyes, are commercially available, such as from Molecular Probes. Inc. (Eugene, Oreg.). The interspecies specificity and approximate affinity of some other non-antibody immunoglobulin-binding proteins bind to segments of a target antibody, such as that of protein A and protein G are known (Langone, Adv. Immunol. 32, 157 (1982); Surolia et al., Trends Biochem. Sci. 7, 74 (1982); Notani et al., J. Histochem. Cytochem. 27, 1438 (1979); Goding, J. Immunol. Meth. 20, 241 (1978); J. Immunol. Meth. 127, 215 (1990); Bjorck et al., J. Immunol. 133, 969 (1984)). [0127]
  • In addition, labeling proteins (goat Fab anti-(mouse Fc), goat Fab anti-(mouse lambda light chain), goat Fab anti-(mouse kappa light chain), protein A, protein G, protein L, lectins, single-chain fragment variable antibodies (ScFv) ) conjugated to the detectable labels of R-phycoerythrin (R-PE), allophycocyanin (APC), tandem conjugates of phycobiliproteins with chemical dyes including several Alexa Fluor dyes, horseradish peroxidase (HRP), [0128] Coprinus cinereus peroxidase, Arthromyces ramosus peroxidase, glucose oxidase and alkaline phosphatase (AP) were or can be prepared by standard means (Haugland et al., Meth. Mol. Biol. 45, 205 (1995); Haugland, Meth. Mol. Biol. 45, 223 (1995); Haugland, Meth. Mol. Biol. 45, 235 (1995); Haugland, Current Protocols in Cell Biol 16.5.1-16.5.22 (2000)). Fusion proteins, such as of protein G or protein A with detectable labels such as luciferin, aequorin, green-fluorescent protein and alkaline phosphatase are also known that are suitable for practice of the invention (Sun et al., J. Immunol. Meth. 152, 43 (1992); Eliasson et al., J. Biol. Chem. 263, 4323 (1988); Eliasson et al., J. Immunol. 142, 575 (1989)).
  • Immunoglobulin heavy and light chains, like most secreted and membrane bound proteins, are synthesized on membrane-bound ribosomes in the rough endoplasmic endoplasmic reticulum where N-linked glycosylation occurs. The specificity of lectins for carbohydrates, including N-linked glycoproteins, is also known (EY laboratories, Inc. Lectin Conjugates Catalog, 1998). [0129]
  • Example 5 Preparation of the Labeled Antibody Immunoglobulin-Binding Protein or the Non-Antibody Immunoglobulin-Binding Peptide and Protein Conjugates while Bound to an Affinity Matrix
  • Unlabeled Fab fragment for goat anti-(mouse Fc) (prepared as in Example 3) was bound to agarose-immobilized mouse IgG for one hour. Following a wash step with bicarbonate buffer, pH 8.3, the complex of immobilized IgG and unlabeled Fab was labeled for one hour at room temperature with the succinimidyl ester of the amine-reactive label. Unconjugated dye was eluted with bicarbonate buffer, and then the covalently labeled Fab fragment was eluted with 50-100 mM glycine/HCl buffer, pH 2.5-2.8. The eluate was collected in 1 mL fractions. The pH of the protein fractions was immediately raised to neutral by addition of 100 μL of either 500 mM phosphate or Tris buffer, pH 7.6, to each 1 mL fraction. Variations of the reagent concentrations, labeling times, buffer composition, elution methods and other variables are possible that can yield equivalent results. Conjugates of the Fab fragment of goat anti-(rabbit Fc) and of protein G and protein A are prepared similarly. [0130]
  • Example 6 Comparison of the Alexa Fluor 488 Dye-Labeled Fab Fragments of Goat Anti-(Mouse Fc) Prepared as in Example 4 and as in Example 5
  • Conjugates of the Fab fragment of goat anti-(mouse Fc) with the [0131] Alexa Fluor 488 succinimidyl ester were separately prepared, as described in Examples 4 and 5. The conjugates had estimated degrees of substitution of ˜1.9 (labeled as in Example 4) and ˜3.0 (labeled as in Example 5), respectively, and virtually identical absorption and emission spectral maxima. When excited at 488 nm, conjugates prepared using the fragment prepared as described in Example 5 were about 3.2-times more fluorescent than using the fragments that were prepared in Example 4 (FIG. 8) as detected by flow cytometry when bound to CD3 on Jurkat T cells. Similar results were observed with other dyes.
  • Example 7 Preparation of a Labeling Protein from Protein G and Albumins
  • Native protein G has a high affinity binding (nanomolar) site for albumins, in particular ovalbumin. Equal weights of protein G and Texas Red ovalbumin (Molecular Probes. Inc.) were dissolved in PBS, pH 7.5. After one hour, the resulting complex was separated on a Sephacryl S-200 Superfine size-exclusion column and analyzed by SDS-PAGE and HPLC. Alternatively, the protein G is combined with a labeled albumin while the protein G is immobilized on any of the several immunoglobulins to which it binds, and the excess labeled albumin is washed away preceding elution of the albumin-labeled protein G complex from the matrix. [0132]
  • Example 8 Preparation of an Immunolabeling Complex on a Very Small Scale
  • Submicrogram quantities of a target-binding antibody were complexed with submicrograms of a labeling protein in varying molar ratios of between about 1:1 and 1:20 to prepare an immunolabeling complex that was suitable for staining a sample. For instance, 0.1 μg of mouse monoclonal anti-α-tubulin in 1 μL PBS with 0.1% BSA was complexed with 0.5 μg of the [0133] Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse Fc) (prepared as in Example 4) or with 0.1 μg of the Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse Fc) (prepared as in Example 5) in 5 μL of PBS for 10 minutes at room temperature. The immunolabeling complex can be used immediately for staining tubulin in fixed-cell preparations (Example 16) or any excess unbound Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse Fc) in the immunolabeling complex can be captured with non-immune mouse IgG (Example 9) for combination with other antibody conjugates, including those of targeting antibodies that have been directly conjugated to other labels. Rabbit antibodies were labeled similarly using labeled goat anti-(rabbit Fc). Labeling of targeting antibodies with a labeled protein A, protein L, protein G, protein G complexed with a labeled albumin, or other immunoglobulin-binding peptides or proteins proceeds similarly. In the case of a mouse (or rat) monoclonal antibody, it is preferred to use a labeled protein that is selective for the specific isotype of the primary antibody (e.g. anti-(mouse IgG1) for a mouse IgG1 isotype primary antibody). Although some cross-reactivity for other mouse (or rat) isotypes was observed using a goat antibody that was selective for mouse IgG1 isotype monoclonal antibodies, routine and optimal use for labeling unmatched mouse isotypes required greater amounts of immunolabeling complexes and was somewhat less reliable.
  • Example 9 Capturing Excess Immunoglobulin-Binding Protein by a Capturing Component
  • Immunolabeling complexes were prepared as described in Example 8. To the immunolabeling complex was added to each [0134] tube 25 μL of a 14.1 mg/mL stock solution of unlabeled mouse IgG to capture excess immunolabeling complexes. As shown in FIG. 1, not all of the immunoglobulin-binding protein was necessarily complexed with the target-binding antibody to form an immunolabeling complex. Consequently, particularly for applications in which labeling complexes of multiple primary antibodies from the same species (e.g. mouse monoclonal antibodies) or crossreacting species (e.g. mouse and human antibodies, FIG. 2, Table 1) were to be used simultaneously or sequentially, it is necessary to quench or otherwise remove any excess immunoglobulin-binding protein by use of a capturing component or by other means to avoid inappropriate labeling of the sample. The most effective capturing component to capture excess immunoglobulin-binding protein is one that contains the binding site of the targeting agent. For instance, whole mouse IgG or mouse serum was shown to be an effective and inexpensive reagent when the immunoglobulin-binding protein was bound to a segment of a mouse monoclonal antibody. The mouse IgG was added in excess to the amount of immunoglobulin-binding protein and incubated for a period of approximately 1-5 minutes, or longer.
  • It is preferred to prepare the immunolabeling complex and then add the capturing component shortly before the experiment. The rapid quenching effect permits this to be done within minutes of performing labeling of the sample by the immunolabeling complex. If desired, the excess capturing component can be removed following labeling of the sample by a simple wash step. Alternatively, fixation of the stained sample by aldehyde-based fixatives or other reagents or methods subsequent to incubation with the immunolabeling complex can provide permanent immobilization of the immunolabeling complex on its target in the sample. As an alternative to adding a soluble capturing component to the immunolabeling complex, the capturing component can be immobilized on an insoluble matrix such as agarose and the immunolabeling complex contacted with that matrix. A preferred matrix when labeling mouse antibodies to mouse antigens is mouse IgG immobilized on agarose. Excess labeled anti-rabbit antibodies can be captured using rabbit IgG that is free in solution or immobilized. Alternatively, the immunolabeling complex can be separated from any capturing component by chromatographic or electrophoretic means. [0135]
  • Example 10 HPLC Analysis of a Labeling Complex
  • In order to analyze the success and extent of complex formation of the labeling protein with the target-binding antibody, size exclusion HPLC of the samples was performed. For instance, a complex of [0136] Alexa Fluor 488 dye-labeled goat Fab anti-(mouse Fc) with a monoclonal mouse anti-tubulin in molar ratios of approximately 1:1, 3:1, 5:1 and 10:1. These were separated by analytical HPLC using a BioSep S-3000 column and eluting with 0.1 M NaPi, 0.1 M NaCl, pH 6.8, at a flow rate of 0.25 mLs/min. An example of the separation using the 5:1 molar ratio (FIG. 6) demonstrates that, using this molar ratio, formation of the labeled complex is essentially quantitative.
  • Example 11 Cross-Reactivity of Goat Fab Anti-(Mouse Fc) to Other Species of IgG
  • Microplates were equilibrated overnight with IgG from a mouse or non-mouse species, and then further blocked with BSA. Variable amounts of the biotinylated Fab fragment of goat anti-(mouse Fc) were added to each well and allowed to bind. After washing, streptavidin-HRP and the Amplex Red peroxidase substrate were added. HRP activity was detected by the addition of H[0137] 2O2 using the Amplex Red Peroxidase Assay Kit (Molecular Probes, Inc., Eugene, Oreg.). Reactions containing 200 μM Amplex Red reagent, 1 U/mL HRP and 1 mM H2O2 (3% solution) in 50 mM sodium phosphate buffer, pH 7.4, were incubated for 30 minutes at room temperature. Fluorescence was measured with a fluorescence microplate reader using excitation at 560±10 nm and fluorescence detection at 590±10 nm. Background fluorescence, determined for a no-H2O2 control reaction, was subtracted from each value (Table 1 and FIG. 2). Table 1 shows that the goat anti-(mouse Fc) antibody because of the highly conserved structure of the Fc region of an antibody it can be used to complex other non-mouse antibodies, including rat, and human antibodies. The goat anti-mouse IgG antibody reaction with mouse antibody was set at 100% and the crossreacting antibodies were expressed as a percentage compared the mouse on mouse data. The data in Table 1 show that the Fab fragment of the goat anti-(mouse Fc) antibody of the current invention does not strongly bind to the goat or sheep Fc domain; however, one skilled in the art could generate antibodies that will react with the goat and sheep Fc domain or the Fc domain of any other species. Biotinylated Fab goat anti-(mouse Fc) was used in this example because it provided a convenient method to quantitate the amount of crossreactivity in a conventional method but it could have been accomplished using a fluorophore Fab labeled goat anti-(mouse Fc). It was demonstrated by HPLC (as in Example 10) that Alexa Fluor 488 dye-labeled goat anti-(rabbit Fc) bound to rabbit primary antibodies.
    TABLE 1
    Cross-reactivity of goat anti-mouse IgG antibody with other non-mouse
    antibodies.
    Species Crossreactivity % Fluorescence
    Mouse ++++ 100
    Rat +++ 80.7
    Human ++ 66.7
    Rabbit + 16.9
    Goat 6.5
    Sheep 5.7
  • Example 12 Determination of the Optimal Molar Ratio of Immunoglobulin-Binding Protein to Target Antibody Using a Microplate Assay
  • To 1.6 μg of mouse monoclonal anti-biotin (MW 145,000) in 8.0 μL PBS was added varying amounts of the [0138] Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse Fc) (MW ˜50,000) (prepared as in Example 4) to form an immunolabeling complex. After equilibration for 20 min, a 100 μL aliquot was added to a 96-well microplate coated with biotinylated BSA. After 30 minutes, the plates were washed and the residual fluorescence was quantitated using a fluorescence microplate reader using excitation at 485+/−10 nm and detecting emission at 530+/−12.5 nm. As shown in FIG. 3, a molar ratio of the Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse Fc) to the anti-biotin between 5 to 20 was sufficient to form appreciably detectable complexes (FIG. 3; fluorescence quantitated, performed in triplicate (circles); control experiments performed but without adding the primary anti-biotin antibody (solid squares)). A molar ratio of about 5 to about 10 was preferred for this pair of immunoglobulin-binding protein and target antibody. This ratio can be varied somewhat to increase or decrease the signal or to affect the consumption of valuable reagents. The weight ratio of immunoglobulin-binding protein to target-binding antibody is particularly affected by the actual molecular weight of the immunoglobulin-binding protein.
  • For instance, equal weights of the dye-labeled goat Fab anti-(mouse Fc) (prepared as in Example 5) and an intact mouse primary antibody, which corresponds to an approximately 3 to 1 molar ratio, usually yields suitable labeling complexes. Fluorescence intensity (or enzymatic activity) of the immunolabeling complex is readily adjusted by a corresponding adjustment of the amount of labeled Fab fragment used. [0139]
  • Similar analyses of the ratio for other labeling proteins (including those of labeled protein A, protein G, protein L, IgG-binding peptides and antibodies to other segments of the primary antibody), and for conjugates of labels other than [0140] Alexa Fluor 488 dye (including enzymes in combination with the appropriate enzyme substrates) are done essentially as described in this example.
  • Example 13 Dissociation Rate of the Immunolabeling Complex
  • A pre-equilibrated immunolabeling complex was prepared from 50 μg of an [0141] Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse Fc) and 15 μg of an anti-biotin monoclonal antibody (mAb). The immunolabeling complex was rapidly diluted with capturing component sufficient to give a 6.2 molar excess over the anti-biotin mAb. At various times, an aliquot was taken and added to a microplate well containing an excess of biotinylated BSA. After 30 minutes, the plates were washed and the remaining fluorescence was quantitated. Displacement of the labeling protein from the target-binding antibody through exchange was measured by any time-dependent decrease in fluorescence in the microplate well. For example the fragments prepared as described in Example 4 had 68 percent fragments bound to the target-binding antibody after 30 minutes compared to 87 percent of bound fragments that were prepared according to Example 5. One hour showed a similar decrease, 56 percent and 68 percent respectively. The labeling protein was shown to undergo a stable interaction with the target-binding antibody, with a lifetime for half exchange under these conditions of 3.5 hours. Dissociation rates were measured for labeling protein prepared according to Example 4 and for labeling protein prepared according to Example 5, demonstrating the greater stability of immunolabeling complexes made using the labeling proteins prepared according to Example 5.
  • Example 14 Protocol for Staining Cultured Cells with a Single Immunolabeling Complex
  • Culturable cells, such as bovine pulmonary artery endothelial cells (BPAEC), were grown on a 22×22 mm glass coverslip. The cells were fixed for 10 minutes using 3.7% formaldehyde in DMEM with fetal calf serum (FCS) at 37° C. The fixed cells were washed 3 times with PBS. The cells were permeabilized for 10 min with 0.02% Triton X-100 in PBS, washed 3× with PBS and blocked for 30 min with 1% BSA in PBS. Variations of the cell type and cell preparation, fixation, and permeabilization methods, including methods for antigen retrieval, are well known to scientists familiar with the art. An immunolabeling complex was prepared as described in Example 8. The immunolabeling complex was added directly to the fixed and permeabilized cells in an amount sufficient to give a detectable signal if there is a binding site for the primary antibody present in the sample. After an incubation period that was typically 10-60 minutes (usually about 15-30 minutes), the cells were washed with fresh medium and the labeling was evaluated by methods suitable for detection of the label. Staining by the immunolabeling complex can be additionally preceded, followed by or combined with staining by additional reagents, such as DAPI, which yields blue-fluorescent nuclei. [0142]
  • Example 15 Protocol for Staining Cultured Cells with Multiple Immunolabeling Complexes
  • Cells were fixed and permeabilized as described in Example 14. Multiple immunolabeling complexes were individually prepared from a variety of labeling proteins, according to the procedure described in Example 8. The multiple immunolabeling complexes were either used individually or sequentially to stain the cells, according to the procedure described in Example 14, or two or more immunolabeling complexes were formed then co-mixed in a single staining solution and used to simultaneously stain the sample. The optimal method for cell fixation and permeabilization and the best ratio for combination of the immunolabeling complexes are typically determined by preliminary experimentation using single immunolabeling complexes or multiple immunolabeling complexes used in combination. A first immunolabeling complex was prepared from an [0143] Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse Fc) and mouse monoclonal anti-α-tubulin, a second immunolabeling complex was prepared from an Alexa Fluor 568 dye-labeled Fab fragment of goat anti-(mouse Fc) and mouse monoclonal anti-vimentin (anti-vimentin was an ascites fluid preparation) and a third immunolabeling complex was prepared from an Alexa Fluor 647 dye-labeled Fab fragment of goat anti-(mouse Fc) and mouse monoclonal anti-cdc6 peptide antibody (Molecular Probes). Aliquots of the three different immunolabeling complexes were combined and used to stain BPAE cells for 30 minutes, washed with fresh medium and observed by fluorescence microscopy using optical filters appropriate for the three dyes. In this example, some cells showed cytoplasmic staining by the anti-vimentin antibody, nuclear staining by the anti-cdc6 peptide antibody and staining of mitotic spindles by the anti-α-tubulin antibody, indicative of a cell in mitosis. Staining by the immunolabeling complexes was additionally preceded, followed by or combined with staining by additional reagents, such as Alexa Fluor 350 phalloidin, which yielded blue-fluorescent actin filaments in the above example.
  • The immunolabeling complexes that are used in combination do not have to be targeted toward antibodies from the same species. For instance, complexes of [0144] Alexa Fluor 488 dye-labeled goat anti-(mouse IgG1 Fc) with a mouse IgG1 monoclonal target-binding antibody and an Alexa Fluor 594 dye-labeled goat anti-(rabbit Fc) with a rabbit primary target-binding antibody can be prepared and used in combined staining protocols.
  • Example 16 Protocol for Staining Tissue with a Single Immunolabeling Complex
  • A mouse intestine cryosection (University of Oregon histology core facility), a cross-section of about 16 μm thickness, was mounted on a slide. The intestine was perfused and fixed with 4% formaldehyde prior to dissection, embedding, and sectioning. The tissue section was rehydrated for 20 minutes in PBS. An immunolabeling complex was prepared as described in Example 8. Briefly, 0.1 μg of mouse monoclonal anti-cdc6 peptide (a nuclear antigen) in 1 μL PBS with 0.1% BSA was complexed with 0.5 μg of the Alexa Fluor 350 dye-labeled Fab fragment of goat anti-(mouse IgG[0145] 1 Fc) (prepared as in Example 4) in 5 μL of PBS for 10 minutes at room temperature. Excess Fab fragment of goat anti-(mouse IgG1 Fc) was captured with 25 μL of a 14.1 mg/mL stock of unlabeled mouse IgG. The tissue was permeabilized with 0.1% Triton X-100 for 10 min.
  • The tissue was washed two times with PBS and was blocked in 1% BSA for 30 min. The immunolabeling complex was added directly to the tissue for 30 minutes and washed three times in PBS. The sample was mounted in Molecular Probes' Prolong antifade mounting medium and observed by fluorescence microscopy using optical filters appropriate for the Alexa Fluor 350 dye. Results showed that the mouse monoclonal anti-cdc6 peptide immunolabeling complex showed specific nuclear labeling in the mouse intestine tissue section. Variations of the tissue type and tissue preparation, fixation and permeabilization methods, mounting methods, including methods for antigen retrieval, are well known to scientists familiar with the art. [0146]
  • Example 17 Staining of a Tissue Target in Combination with Tyramide Signal Amplification (TSA)
  • Mouse brain cryosections were labeled with a pre-formed complex of horseradish peroxidase (HRP)-labeled goat anti-(mouse IgG[0147] 1 Fc) antibody and a mouse IgG1 monoclonal anti-(glial fibrillary acidic protein (GFAP)) prepared essentially as in Example 8 using a molar ratio of labeling protein to monoclonal antibody of 3. Staining of the mouse tissues was essentially as in Example 16. The staining localization and intensity was compared to that of (a) goat anti-mouse IgG HRP conjugate and mouse anti-GFAP, (b) the Alexa Fluor 488 dye-labeled Fab fragment of goat anti-mouse IgG1 Fc) antibody complex of mouse anti-GFAP, (c) Alexa Fluor 488 goat anti-mouse IgG secondary antibody and mouse anti-GFAP, and (d) a direct conjugate of the Alexa Fluor 488 dye with mouse anti-GFAP. The HRP-conjugated probes were incubated with Alexa Fluor 488 tyramide using TSA Kit #2 (Molecular Probes, Inc.) according to standard procedures. The tissue staining patterns in each case were similar and consistent with the expected staining pattern of mouse anti-GFAP and staining was essentially free of nonspecific background. The relative fluorescence intensities of staining measured by digital imaging were sequentially: 541 relative intensity units for the HRP-goat anti-(mouse IgG1 Fc) complex of mouse anti-GFAP and (using the combinations indicated by the letters above): (a) 539, (b) 234, (c) 294, and (d) 255 relative intensity units.
  • Example 18 Staining of Live Cells by Multiple Immunolabeling Complexes
  • A first immunolabeling complex was prepared from an [0148] Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse IgG1 Fc) and mouse monoclonal anti-(human CD8), a second immunolabeling complex was prepared from an R-phycoerythrin-conjugated Fab fragment of goat anti-(mouse IgG1 Fc) and mouse anti-(human CD3), and a third immunolabeling complex was prepared from an Alexa Fluor 647 dye-labeled Fab fragment of goat anti-(mouse IgG1 Fc) and mouse anti-(human CD4). The complexes were prepared as described in Example 8 and were each blocked with 20 μg (1.3 μL of 14.1 μg/mL) of mouse IgG for 10 minutes at room temperature. The first immunolabeling complex was added to 100 μL of whole blood and incubated for 15 min. The cells were washed with PBS and 280.5 μL of the second immunolabeling complex was added and incubated for 15 min. The cells were again washed, and 46.2 μL of the third labeling complex was added and incubated for 15 min. After the final incubation, the red blood cells were lysed with cell-lysis buffer. The cells were resuspended in 1% formaldehyde/PBS and analyzed on a FACS Vantage flow cytometer using a 488 nm argon-ion laser for excitation of the first and second immunolabeling complexes and a 633 nm red He—Ne laser for excitation of the third immunolabeling complex (FIGS. 5a, 5 b). The emission band pass filters used for selective detection of the dyes are 525+/−10 nm for the Alexa Fluor 488 (CD8), 585+/−21 nm for R-PE (CD3) and 675+/−10 nm for the Alexa Fluor 647 dye (CD4). FIGS. 5a and 5 b show that the instant invention can be used in a 3-color immunophenotyping experiment using peripheral blood lymphocytes. CD3-positive T cells were stained with the R-phycoerythrin-conjugated Fab fragment of goat anti-(mouse Fc) and mouse anti-human CD3), upper left (UL) quadrant, FIG. 5a. CD4-positive cells, a T cell subset, are identified using Alexa Fluor 647 dye-labeled Fab fragment of goat anti-(mouse IgG1 Fc) and mouse anti-(human CD4), UL quadrant, FIG. 5b and CD8-positive T cells, a T cell subset, were identified using Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse IgG1 Fc) and mouse monoclonal anti-(human CD8), lower right (LR) quadrant, FIG. 5b.
  • Exposed antigens of live cells, including cultured cells and cells from biological fluids such as blood and cerebrospinal fluid can be simultaneously or sequentially stained by combinations of immunolabeling complexes, including antibodies to the same target labeled with two or more separately detectable immunoglobulin-binding proteins. [0149]
  • Example 19 The Dye-Labeled Fab Fragment of Goat Anti-(Mouse Fc) can be Utilized for the Combinatorial Labeling of Primary Antibodies, to Generate a Multitude of Colored Targets
  • A first immunolabeling complex was made by combining 2.5 [0150] μg Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse IgG1 Fc) with 0.5 μg mouse anti-human CD3 (Caltag at 200 μg/mL), according to the procedure described in Example 4. A second immunolabeling complex was made by combining 5.0 μg Alexa Fluor 647 dye-labeled Fab fragment of goat anti-(mouse IgG1 Fc) with 0.5 μg mouse anti-human CD3, according to the procedure in Example 4. Each complex was separately incubated at room temperature for 5 minutes, and each complex was then separately combined with an excess of mouse IgG (14.1 mg/mL) for 5 min at room temperature to capture excess unbound dye-labeled Fab fragments. The two immunolabeling complexes were then added in different percentage combinations (see Table 2) to 100 μL of washed heparinized blood. The cells were incubated with the respective combinations of complexes for 20 min on ice. The red blood cells were then lysed with a cell-lysis buffer. The cells were resuspended in 1% formaldehyde/PBS and analyzed on a FacVantage flow cytometer using a 488 nm argon 633 HeNe laser for excitation and a 530+/−10 nm band pass emission filter (FL1), and a 640 long pass filter (FL4). Five samples of different combined percentages (Table 2) were compared by flow cytometry, with signals being collected in FL1 and FL4. To determine the percentage of cells detected with each type of emission, the FL1 and FL4 intensities for each percentage combination were normalized by dividing the FL1 and FL4 channel intensities for such combinations by the intensities of the 100% Alexa Fluor 488 dye- and 100% Alexa Fluor 647 dye-labeled cells, respectively.
    TABLE 2
    Theoretical versus recovered dye-labeled Fab fragment of goat anti-(mouse
    IgG1 Fc) combinatorial experiment.
    Recovered
    percentage of
    measured cells
    Experimentally mixed Recovered percentage Experimentally mixed labeled with Alexa
    percentage of cells of measured cells percentage of cells Fluor 647 dye-
    labeled with Alexa labeled with Alexa labeled with Alexa labeled Fab
    Fluor
    488 dye-labeled Fluor 488 dye-labeled Fluor 647 dye-labeled fragment of goat
    Fab fragment of goat Fab fragment of goat Fab fragment of goat anti-(mouse IgG1
    anti-(mouse IgG1 Fc) anti-(mouse IgG1 Fc) anti-(mouse IgG1 Fc) Fc)
    100% 100% 0% 0%
    75% 81% 25% 14%
    50% 63% 50% 38%
    25% 35% 75% 73%
    0% 0% 100% 100%
  • Example 20 The Immunolabeling Complex can be used to Detect Antigens on a Western Blot
  • Bovine heart mitochondria were isolated (Hanson et al., Electrophoresis 22, 950 (2001)). The isolated mitochondria were resuspended to ˜10 mg/mL in 100 mM Tris-HCl, pH 7.8, 1 mM phenylmethylsulfonyl fluoride (a protease inhibitor), 2% SDS and insoluble material was removed by centrifugation for 10 minutes at 10,000×g in a tabletop centrifuge. The protein concentration of the lysate was checked by the BCA assay (Pierce, Rockford, Ill.). Samples for gel electrophoresis were prepared by mixing lysate, water, and loading buffer to the appropriate concentrations (final concentration of loading buffer in samples: 58 mM Tris/HCl, 10% glycerol, 2% SDS, 0.02 mg/mL bromphenol blue, 50 mM DTT, pH 8.6). The samples were then heated to 90° C. for 5 minutes before loading on the gel and separated on a 13% SDS-PAGE gel. Two-fold serial dilution of the extracts ranging from 8 μg of extract down to 0.03 μg were loaded on the SDS-PAGE gel. The proteins were transferred to PVDF membrane for 1.5 hours using a semi-dry transfer system according to manufacturer's directions (The W.E.P. Company, Concord, Calif.). The PVDF membrane was blocked for l hour in 5% milk. [0151]
  • Immunolabeling complexes were made with mouse monoclonal antibodies that recognize two different mitochondrial proteins. [0152] Alexa Fluor 647 dye-labeled Fab fragment of goat anti-(mouse IgG1 Fc) (5 μL of a 1 mg/mL stock, prepared as in Example 4) was incubated with 21 μL (0.88 mg/mL) mouse anti-(CV-alpha) and Alexa Fluor 488 dye-labeled Fab fragment of goat anti-(mouse IgG1 Fc) (5 μL of a 1 mg/mL stock, prepared as in Example 4) was incubated with 19 μL (0.88 mg/mL) mouse anti-(CIII-core2) (Molecular Probes, Eugene, Oreg.). Following a 30 minute incubation, 25 μL of a 14.1 mg/mL stock of unlabeled mouse IgG was added to each tube. The immunolabeling complexes were then mixed together and brought up to 5 mL in 5% milk. The blot was incubated with the mixture of immunolabeling complexes for 1 hour at room temperature. The blot was washed twice for 5 seconds each with PBST (PBS with 0.1% Tween) and once with PBST for 15 minutes. The blot was air dried and imaged on an EG&G Wallac Imager with the appropriate filters. The Western blot revealed two distinct bands of the appropriate molecular weight. The Western blot also showed that no cross-labeling of the antibodies occurred and the detection limit was 125 ng.
  • Example 21 High-Throughput Screening of Hybridomas for Identifying High Affinity and High IgG Producers
  • Microplate wells containing both a fluorescent labeled antigen of one fluorescent color label and fluorescently labeled Fab fragments of goat anti-(mouse Fc) of a different fluorescent color made by the method described in Example 4 and 5. Hybridoma supernatant is harvested and added to the wells. If the hybridoma are producing the desired antibody, i.e. antibodies that bind to the labeled antigen, polarization of the florescence corresponding to the labeled antigen will allow visualization of those wells containing antigen specific antibody. In addition, the amount of IgG which the hybridomas produce, can be simultaneously identified by polarization of the fluorescence corresponding to the labeled Fab fragments. This method thus allows for both quantitation of the amount of antibody present in a specific amount of hybridoma supernatant and the affinity of the monoclonal antibodies for the antigen. [0153]
  • The reagents employed in the preceding examples are commercially available or can be prepared using commercially available instrumentation, methods, or reagents known in the art or whose preparation is described in the examples. It is evident from the above description and results that the subject invention is greatly superior to the presently available methods for determining the presence of a target in a biological sample. The subject invention overcomes the shortcomings of the currently used methods by allowing small quantities of antibodies to be labeled and in unlimited media while maintaining specificity and sensitivity. The examples are not intended to provide an exhaustive description of the many different embodiments of the invention. Thus, although the forgoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, those of ordinary skill in the art will realize readily that many changes and modifications can be made thereto without departing from the spirit or scope of the appended claims. [0154]
  • All publications and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference. [0155]

Claims (83)

What is claimed is:
1. A method for determining the presence of a desired target in a biological sample, said method comprising:
a) combining an immunolabeling mixture with said sample wherein said immunolabeling mixture comprises a target-binding antibody bound to a labeling protein, wherein said labeling protein comprises a monovalent antibody fragment or a non-antibody peptide or protein, wherein said labeling protein is covalently linked to one or more labels; and
b) detecting the presence or absence of said label whereby the presence or absence of said target is determined.
2. The method according to claim 1, wherein said labeling mixture contains at least one target-binding antibody bound by said labeling protein wherein said target-binding antibody is a single chain fragment variable, and said labeling protein binds selectively and with high affinity to a selected peptide linker region of the target-binding antibody.
3. The method according to claim 1, wherein the labeling mixture contains two or more target-binding antibodies bound by labeling proteins, wherein one of said target-binding antibodies is bound by a first labeling protein and another said target-binding antibody is bound by a second labeling protein.
4. The method according to claim 1, wherein said target-binding antibody has two or more different labeling proteins bound to the antibody and at least one labeling protein is detectably distinct from other labeling proteins.
5. The method according to claim 1, wherein said non-antibody protein is selected from the group consisting of a protein G, a protein A, a protein L, a lectin, and a protein G bound to albumin, wherein said albumin is covalently linked to one or more labels and albumin is selected from the group consisting of human albumin, bovine serum albumin, and ovalbumin.
6. The method according to claim 1, wherein said monovalent antibody fragment is a Fab or Fab′ fragment and is selected from the group consisting of anti-Fc antibody fragment, anti-kappa light chain antibody fragment, anti-lambda light chain antibody fragment, and a single chain variable protein fragment.
7. The method according to claim 6, wherein said antibody fragment is derived from a monoclonal antibody or polyclonal antibody.
8. The method according to claim 7, wherein said polyclonal antibody is a rabbit or a goat polyclonal antibody.
9. The method according to claim 7, wherein said monoclonal antibody is classified as mouse IgM, IgG1, IgG2a, IgG2b, or IgG3 antibody, or an equivalent thereof.
10. The method according to claim 1, wherein said label is selected from the group consisting of a fluorescent dye, a phosphorescent dye, a tandem dye, a particle, an electron transfer agent, biotin and a radioisotope.
11. The method according to claim 10, wherein said fluorescent dye is selected from the group consisting of a fluorescent sulfonated xanthene dye, a fluorescent sulfonated cyanine dye, and a fluorescent phycobiliprotein dye.
12. The method according to claim 10, wherein said tandem dye is selected from the group consisting of sulfonated cyanine-phycobiliprotein tandem dye and sulfonated xanthene-phycobiliprotein tandem dye.
13. The method according to claim 1, wherein said biological sample comprises eukaryotic or prokaryotic cells, cellular extract, subcellular component, tissue culture, tissue, a bodily fluid, or a portion or combination thereof.
14. The method according to claim 1, wherein said target-binding antibody binds to an antigenic determinate present in the biological sample of claim 13.
15. The method according to claim 13, wherein said sample is immobilized on a surface.
16. The method according to claim 13, wherein said sample is in a gel or on a blot or in an array.
17. The method according to claim 1, wherein the amount of target-binding antibody is about 5 micrograms to about 0.001 micrograms.
18. The method according to claim 1, further comprising binding of unbound labeling protein prior to combining labeling mixture with sample, wherein a capture component is added to the labeling mixture and binding said unbound labeling protein.
19. The method of claim 18, wherein said capture component is an antibody or an antibody fragment, and is optionally attached to a microsphere or to agarose.
20. The method according to claim 1, wherein said labeling mixture contains at least one target-binding antibody bound to a labeling protein and
i) the labeling protein is a fragment of a rabbit or goat polyclonal antibody or of a mouse IgG1, IgG2a, IgG2b, IgG3 antibody,
ii) the labeling protein is an anti-Fc antibody fragment that is a monovalent Fab or Fab′ fragment, wherein the labeling protein is covalently labeled with one or more labels that are selected from the group consisting of fluorescent sulfonated xanthene dye, fluorescent sulfonated cyanine dye, fluorescent phycobiliprotein dye, sulfonated cyanine-phycobiliprotein tandem dye, and sulfonated xanthene-phycobiliprotein tandem dye, which labels are optionally the same or different; and
the capture component is an antibody or an antibody fragment, which is optionally attached to a microsphere or to agarose.
21. A method for determining the presence of a desired target in a biological sample, said method comprising:
a) combining an immunolabeling mixture with said sample wherein said immunolabeling mixture comprises a first target-binding antibody bound to a first labeling protein and a second target-binding antibody bound to a second labeling protein, wherein said first and second labeling proteins comprise a monovalent antibody fragment or a non-antibody peptide or protein that are detectably distinct, and wherein said labeling proteins are covalently linked to one or more labels; and
b) detecting the presence or absence of said labels whereby the presence or absence of said target is determined.
22. The method according to claim 21, wherein said first labeling protein is a non-antibody peptide or protein and said second labeling protein is a non-antibody peptide or protein.
24. The method according to claim 22, wherein said first and second non-antibody proteins are independently selected from the group consisting of a protein G, a protein A, a protein L, a lectin, and a G protein bound to albumin, wherein said albumin is covalently linked to one or more labels and albumin is selected from the group consisting of human albumin, bovine serum albumin, and ovalbumin.
25. The method of claim 21, wherein said first labeling protein is a first monovalent antibody fragment and said second labeling protein is a second monovalent antibody fragment.
26. The method according to claim 25, wherein said first and second monovalent antibody fragments are independently Fab or Fab′ fragments and are selected from the group consisting of anti-Fc antibody fragment, anti-kappa light chain antibody fragment, anti-lambda light chain antibody fragment, and a single chain variable protein fragment.
27. The method according to claim 25, wherein said first and second antibody fragments are derived from a monoclonal antibody or polyclonal antibody.
28. The method according to claim 27, wherein said polyclonal antibody is a rabbit or a goat polyclonal antibody.
29. The method according to claim 27, wherein said monoclonal antibody is classified as mouse IgM, IgG1, IgG2a, IgG2b, or IgG3 antibody, or an equivalent thereof.
30. The method according to claim 21, wherein said labels are selected from the group consisting of a fluorescent dye, a phosphorescent dye, a tandem dye, a particle, an electron transfer agent, biotin and a radioisotope.
31. The method according to claim 30, wherein said fluorescent dye is selected from the group consisting of a fluorescent sulfonated xanthene dye, a fluorescent sulfonated cyanine dye, and a fluorescent phycobiliprotein dye.
32. The method according to claim 30, wherein said tandem dye is selected from the group consisting of sulfonated cyanine-phycobiliprotein tandem dye and sulfonated xanthene-phycobiliprotein tandem dye.
33. The method according to claim 21, wherein said biological sample comprises eukaryotic or prokaryotic cells, cellular extract, subcellular component, tissue culture, tissue, a bodily fluid, or a portion or combination thereof.
34. The method according to claim 33, wherein said target-binding antibody binds to an antigenic determinate present in said biological sample.
35. The method according to claim 34, wherein the amount of target-binding antibody is about 5 micrograms to about 0.001 micrograms.
36. The method of claim 21, wherein said sample is immobilized on a solid surface.
37. The method of claim 21, wherein said sample is in a gel, or on a blot or in an array.
38. The method according to claim 21, further comprising binding of unbound labeling protein prior to combining labeling mixture with sample, wherein a capture component is added to the labeling mixture binding said unbound labeling protein.
39. The method of claim 38, wherein said capture component is an antibody or an antibody fragment, and is optionally attached to a microsphere or to agarose.
40. A method according to claim 21 wherein said labeling mixture contains at least two target-binding antibodies each bound by at least one labeling protein; and
a) said first and second labeling proteins are independently a fragment of a rabbit or goat polyclonal antibody or a mouse IgG1, IgG2a, IgG2b, IgG3 antibody,
b) said first and second labeling proteins are an anti-Fc antibody fragment that is a monovalent Fab or Fab′ fragment, wherein said first and second labeling proteins are covalently labeled with one or more labels that are independently selected from the group consisting of fluorescent sulfonated xanthene dye, fluorescent sulfonated cyanine dye, fluorescent phycobiliprotein dye, sulfonated cyanine-phycobiliprotein tandem dye, and sulfonated xanthene-phycobiliprotein tandem dye, which labels are optionally the same or different; and
the capture component is an antibody or an antibody fragment, which is optionally attached to a microsphere or to agarose.
41. A method for determining the presence of a desired target in a biological sample, said method comprising:
a) combining an immunolabeling mixture with said sample wherein said immunolabeling mixture comprises a target-binding antibody bound to a labeling protein, wherein said labeling protein comprises a monovalent antibody fragment or a non-antibody peptide or protein, and wherein said labeling protein is covalently linked to one or more labels; and
b) adding an enzymatic substrate, which is selected from the group consisting of a chromogenic, a fluorogenic and chemiluminescent substrate to the sample combined with said immunolabeled mixture; and
c) detecting the presence or absence of said label whereby the presence or absence of said target is determined.
42. The method according to claim 41, wherein said substrate is a peroxidase substrate, a luciferase substrate, a phosphatase substrate, a glycosidase substrate or a luciferase substrate..
43. The method according to claim 42, wherein said substrate is activated by an enzyme, which is selected from the group consisting of peroxidase, phosphatase, glycosidase, and luciferase.
44. The method according to claim 43, wherein said peroxidase is horseradish peroxidase, Coprinus cinereus peroxidase, or Arthromyces ramosus peroxidase.
45. The method according to claim 43, wherein said phosphatase is alkaline phosphatase.
46. The method according to claim 43, wherein said glycosidase is β-galactosidase or β-glucuronidase.
47. The method according to claim 43, wherein said luciferase is protein A luciferase or protein G luciferase.
48. An isolated immunolabeling complex consisting essentially of:
a target-binding antibody bound by one or more labeling proteins that is a non-antibody peptide or protein, or a monovalent antibody fragment, wherein said labeling protein(s) bind selectively and with high affinity to a selected region of the target-binding antibody, and wherein said labeling protein is covalently linked to one or more labels.
49. The immunolabeling complex according to claim 48, wherein at least one labeling protein is detectably distinct from the other labeling proteins.
50. An isolated immunolabeling complex consisting essentially of:
a target-binding antibody bound by one or more monovalent labeling proteins that is a non-antibody immunoglobulin-binding peptide or protein, or is a monovalent antibody fragment, wherein said labeling protein binds selectively and with high affinity to a selected region of the target-binding antibody, and where each labeling protein is covalently labeled with an enzyme.
51. An isolated immunolabeling complex consisting essentially of:
a target-binding antibody bound by one or more monovalent labeling proteins that is a non-antibody immunoglobulin-binding peptide or protein, or is a monovalent antibody fragment, wherein said labeling protein binds selectively and with high affinity to a selected region of the target-binding antibody, and where each labeling protein is covalently labeled with a hapten.
52. An isolated monovalent labeling protein consisting essentially of a monovalent fragment generated from an antibody and capable of binding selectively and with high affinity to a selected region of a second antibody, wherein said monovalent protein is covalently labeled with one or more labels, and which labels are optionally the same or different.
53. An isolated labeling mixture comprising one or more immunolabeling complexes, wherein at least one immunolabeling complex consists essentially of:
a target-binding antibody bound by a labeling protein, that is a non-antibody peptide or protein, or is a monovalent antibody fragment, wherein said labeling protein binds selectively and with high affinity to a selected region of the target-binding antibody, and the labeling protein is covalently labeled with one or more labels, which labels are optionally the same or different.
54. The isolated labeling mixture according to claim 53, further comprising a capture component that is an antibody or antibody fragment and is optionally attached to a microsphere or to agarose.
55. An isolated labeling mixture comprising one or more immunolabeling complexes, wherein at least one immunolabeling complex consist essentially of a target-binding antibody bound by a labeling protein that is a protein G complexed with albumin, wherein said albumin is covalently labeled with one or more labels, and which labels are optionally the same or different.
56. The isolated labeling mixture according to claim 55, wherein said albumin is human albumin, bovine serum albumin, or ovalbumin.
57. An isolated protein G complex consisting essentially of protein G and albumin, wherein said albumin is covalently labeled with one or more labels, which labels are optionally the same or different.
58. The isolated protein G complex according to claim 57, wherein said albumin is human albumin, bovine serum albumin, or ovalbumin.
59. A kit for preparing an immunolabeling complex, said kit comprising:
a) a labeling mixture consisting essentially of one or more monovalent antibody fragments that bind selectively and with high affinity to a specific region of one or more target-binding antibodies, where each fragment is covalently labeled with one or more labels, and which said labels are optionally the same or different and a capture component to which the antibody fragments bind, that is optionally attached to a microsphere or to agarose.
60. A kit for preparing an immunolabeling complex, said kit comprising:
a) a protein G complex with albumin, wherein said albumin is covalently labeled with one or more labels, and which labels are optionally the same or different; and
b) a capture component to which the protein G complexed with albumin binds, wherein said capture component is optionally attached to a microsphere or to agarose.
61. A kit for immunolabeling a desired target in a biological sample, said kit comprising:
a) a labeling mixture of one or more immunolabeling complexes, wherein at least one immunolabeling complex comprises a target-binding antibody bound by a protein G complexed with albumin, wherein said albumin is covalently labeled with one or more labels, and which detectable moieties are optionally the same or different; and
b) a buffer.
62. The kit according to claim 61, wherein said albumin is human albumin, bovine serum albumin, or ovalbumin.
63. A kit for immunolabeling a target in a sample, said kit comprising:
a) a labeling mixture of one or more immunolabeling complexes, wherein at least one immunolabeling complex comprises a target-binding antibody bound by a labeling protein that is a non-antibody peptide or protein, or a monovalent antibody fragment, wherein said labeling protein binds selectively and with high affinity to a selected region of the target-binding antibody, and where the labeling protein is covalently labeled with one or more labels, which directly detectable moieties are optionally the same or different; and
b) a buffer.
64. An isolated monovalent fragment for labeling antibodies made by the process comprising the steps of:
a) generating a labeling antibody against an antigen, where said antigen is a Fc region of an antibody from a different species;
b) digesting said labeling antibody to generate multiple monovalent fragments;
c) covalently labeling said monovalent fragments with one or more labels, where said labels on each monovalent fragment are optionally the same or different; and
d) isolating said monovalent fragments before or after covalent labeling.
65. The isolated monovalent fragment for labeling target-binding antibodies made by the process according to claim 64, wherein said species is selected from the group consisting of mouse, rabbit and goat.
66. The isolated monovalent fragment for labeling target-binding antibodies made by the process according to claim 64, wherein said labeling antibody is digested with papain to yield multiple Fab fragments.
67. The isolated monovalent fragment for labeling target-binding antibodies made by the process according to claim 64, wherein said fragments are covalently labeled with one or more directly detectable fluorescent dyes, phosphorescent dyes, dye tandems, fluorescent proteins, particles, or electron transfer agents, or combinations thereof.
68. The isolated monovalent fragment for labeling target-binding antibodies made by the process according to claim 64, wherein;
a) before said fragments are covalently labeled, said fragments are immobilized by binding to one or more complementary antibodies, where such binding protects the binding region of such fragments;
b) after said fragments are covalently labeled, isolating said monovalent fragments by dissociating such fragments from such complementary antibodies.
69. An isolated monovalent fragment for labeling target-binding antibodies made by the process comprising the steps of:
a) generating a labeling antibody against an antigen, where said antigen is a selected peptide linker region of a single-chain variable protein fragment;
b) digesting said labeling antibody to generate multiple monovalent fragments;
c) covalently labeling said monovalent fragments with one or more labels, where said labels on each monovalent fragment are optionally the same or different; and
d) isolating said monovalent fragments before or after covalent labeling.
70. An isolated immunolabeling complex made by a process comprising:
a) combining, in vitro, one or more target-binding antibodies with an amount of labeling protein in excess of a ratio of at least one labeling protein to each target-binding antibody, under conditions suitable for the labeling protein to bind to the target-binding antibodies, and where each labeling protein is a non-antibody peptide or protein, or is a monovalent antibody fragment, wherein said labeling protein binds selectively and with high affinity to a selected region of said target-binding antibody to form an immunolabeling complex, and each labeling protein is covalently labeled with one or more labels, where the said labels on each labeling protein are optionally the same or different;
b) isolating said immunolabeling complex after said labeling protein binds to the target-binding antibodies.
71. The isolated immunolabeling complex made by the process according to claim 70, wherein said target-binding antibodies that are combined with the labeling protein are present in a heterogeneous protein mixture.
72. The isolated immunolabeling complex made by the process according to claim 70, wherein said heterogeneous protein mixture is ascites fluid or hybridoma culture supernatant.
73. The isolated immunolabeling complex made by the process according to claim 70, wherein said labeling protein is in excess of a ratio of 3 moles of labeling protein for each mole of target-binding antibody.
74. The isolated immunolabeling complex made by the process according to claim 70, wherein at least one labeling protein is a monovalent antibody fragment, and said monovalent fragment is covalently labeled with one or more fluorescent dyes, chemiluminescent dyes, tandem dyes, fluorescent proteins, or combinations thereof.
75. The isolated immunolabeling complex made by the process according to claim 70, wherein said target-binding antibody binds to an antigenic determinate present in a biological sample.
76. The isolated immunolabeling complex made by the process according to claim 70, wherein at least one labeling protein is a monovalent fragment that has been made by the process consisting essentially of:
a) generating a labeling antibody against an antigen, where said antigen is a Fc region of an antibody from a different species;
b) digesting said labeling antibody to generate multiple monovalent fragments;
c) covalently labeling said monovalent fragments with one or more labels, where said labels on each monovalent fragment are optionally the same or different; and
d) isolating said monovalent fragments before or after covalent labeling.
77. The isolated monovalent fragment made by the process according to claim 76, wherein said labeling protein that is not bound to the target-binding antibodies are bound to a capture component, which is optionally attached to a microsphere or to agarose.
78. The monovalent fragment made by the process according to claim 76, wherein the step of covalently labeling said monovalent fragments is accomplished by immobilizing said monovalent fragments by binding to one or more complementary antibodies, where such binding protects the binding region of such fragments; and
wherein the step of isolating said monovalent fragments is accomplished by dissociating such fragments from such complementary antibodies after covalent labeling.
79. The isolated monovalent fragment made by the process according to claim 76, wherein said species is selected from the group consisting of mouse, rabbit or goat.
80. The isolated monovalent fragment made by the process according to claim 76, wherein said labeling antibody is digested with papain to yield multiple Fab fragments.
81. The isolated immunolabeling complex made by the process according to claim 70, wherein the target-binding antibody is a single-chain variable protein fragment wherein at least one labeling protein is a monovalent fragment that has been made by the process consisting essentially of:
a) generating a labeling antibody against an antigen, wherein said antigen is a selected peptide linker region of said single-chain variable protein fragment;
b) digesting said labeling antibody to generate multiple monovalent fragments;
c) covalently labeling said monovalent fragments with one or more labels, where said labels on each monovalent fragment are optionally the same or different; and
d) isolating said monovalent fragments before or after covalent labeling.
82. The isolated monovalent fragment for labeling antibodies made by the process according to claim 81, wherein said labeling antibody is digested with papain to yield multiple Fab fragments.
83. A method according to claim 1, further comprising stabilizing binding of said target-binding antibody to said labeling protein.
84. A kit for preparing labeling proteins, said kit comprising:
a) a monovalent antibody fragment that binds selectively and with high affinity to a specific region of one or more target-binding antibodies,
b) a chemically reactive dye; and
c) an immobilization matrix comprising the binding domain of the labeling protein.
US10/118,204 2001-10-12 2002-04-05 Antibody complexes and methods for immunolabeling Expired - Lifetime US8323903B2 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
US10/118,204 US8323903B2 (en) 2001-10-12 2002-04-05 Antibody complexes and methods for immunolabeling
EP02768949A EP1442302B1 (en) 2001-10-12 2002-10-02 Antibody complexes and methods for immunolabeling
JP2003533851A JP2006513398A (en) 2001-10-12 2002-10-02 Antibody conjugates and methods for immunolabeling
CA002462280A CA2462280C (en) 2001-10-12 2002-10-02 Antibody complexes and methods for immunolabeling
EP09163588A EP2113773B1 (en) 2001-10-12 2002-10-02 Antibody complexes
AT02768949T ATE436022T1 (en) 2001-10-12 2002-10-02 ANTIBODY COMPLEXES AND IMMUNE LABELING PROCEDURES
EP10181177A EP2259067B1 (en) 2001-10-12 2002-10-02 Methods for immunolabeling
PCT/US2002/031416 WO2003030817A2 (en) 2001-10-12 2002-10-02 Antibody complexes and methods for immunolabeling
DE60232904T DE60232904D1 (en) 2001-10-12 2002-10-02 ANTIBODY COMPLEXES AND IMMUNE MARKING METHOD
GB0410515A GB2397302B (en) 2001-10-12 2002-10-02 Antibody complexes and methods for immunolabeling
US10/666,291 US20070269902A1 (en) 2001-10-12 2003-09-17 Antibody complexes and methods for immunolabeling
JP2007083130A JP4977512B2 (en) 2001-10-12 2007-03-27 Antibody conjugates and methods for immunolabeling
US13/610,009 US8535894B2 (en) 2001-10-12 2012-09-11 Antibody complexes and methods for immunolabeling

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US32906801P 2001-10-12 2001-10-12
US36941802P 2002-04-01 2002-04-01
US10/118,204 US8323903B2 (en) 2001-10-12 2002-04-05 Antibody complexes and methods for immunolabeling

Related Child Applications (5)

Application Number Title Priority Date Filing Date
PCT/US2002/031416 Continuation WO2003030817A2 (en) 2001-10-12 2002-10-02 Antibody complexes and methods for immunolabeling
PCT/US2002/031416 Continuation-In-Part WO2003030817A2 (en) 2001-10-12 2002-10-02 Antibody complexes and methods for immunolabeling
US10/467,550 Continuation US20050069962A1 (en) 2001-10-12 2002-10-02 Antibody complexes and methods for immunolabeling
US10/467,550 Continuation-In-Part US20050069962A1 (en) 2001-10-12 2002-10-02 Antibody complexes and methods for immunolabeling
US10467550 Continuation 2002-10-02

Publications (2)

Publication Number Publication Date
US20030073149A1 true US20030073149A1 (en) 2003-04-17
US8323903B2 US8323903B2 (en) 2012-12-04

Family

ID=27382120

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/118,204 Expired - Lifetime US8323903B2 (en) 2001-10-12 2002-04-05 Antibody complexes and methods for immunolabeling
US10/666,291 Abandoned US20070269902A1 (en) 2001-10-12 2003-09-17 Antibody complexes and methods for immunolabeling

Family Applications After (1)

Application Number Title Priority Date Filing Date
US10/666,291 Abandoned US20070269902A1 (en) 2001-10-12 2003-09-17 Antibody complexes and methods for immunolabeling

Country Status (8)

Country Link
US (2) US8323903B2 (en)
EP (3) EP2113773B1 (en)
JP (2) JP2006513398A (en)
AT (1) ATE436022T1 (en)
CA (1) CA2462280C (en)
DE (1) DE60232904D1 (en)
GB (1) GB2397302B (en)
WO (1) WO2003030817A2 (en)

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050069962A1 (en) * 2001-10-12 2005-03-31 Archer Robert M Antibody complexes and methods for immunolabeling
US20050214882A1 (en) * 2004-03-25 2005-09-29 Ez Bio Inc. Reagents, methods and kits for the universal rapid immuno-detection
US20060115860A1 (en) * 2004-11-26 2006-06-01 Cabrera Robert M Immunologic assay for detection of autoantibodies to folate binding protein
US20070154958A1 (en) * 2005-10-13 2007-07-05 Aureon Laboratories, Inc. Multiplex in situ immunohistochemical analysis
US20070190597A1 (en) * 2006-02-10 2007-08-16 Invitrogen Corporation Oligosaccharide modification and labeling of proteins
WO2008020823A2 (en) * 2005-05-23 2008-02-21 University Of Hawaii Cooperative reporter systems, components, and methods for analyte detection
US20080118916A1 (en) * 2006-11-16 2008-05-22 General Electric Company Sequential analysis of biological samples
US20080118934A1 (en) * 2006-11-16 2008-05-22 General Electric Company Sequential analysis of biological samples
US20080233660A1 (en) * 2007-03-23 2008-09-25 Mu Bioteknik Ab Solid phase labeling method
WO2008133728A2 (en) * 2006-11-16 2008-11-06 General Electric Company Sequential analysis of biological samples with intermediate bleaching of fluorescence detector
US20080286881A1 (en) * 2007-05-14 2008-11-20 Apel William A Compositions and methods for combining report antibodies
US20080286275A1 (en) * 2004-07-08 2008-11-20 Riccardo Cortese Antigen Binding Proteins Directed Against Scavenger Receptor B1 that Inhibit Hcv Replication
US20090035216A1 (en) * 2007-08-03 2009-02-05 Biomonitor Aps Method for determining in vivo biopharmaceutical concentration or bioavailability
US20090263879A1 (en) * 2008-04-21 2009-10-22 Chiu Chin Chang Immunoassay for specific determination of S-adenosylmethionine and analogs thereof in biological samples
US20100009342A1 (en) * 2005-09-06 2010-01-14 Life Technologies Corporation Control of chemical modification
US20100034737A1 (en) * 2006-09-28 2010-02-11 Pierre-Yves Dietrich Junctional adhesion molecule-c (jam-c) binding compounds and methods of their use
US20100120043A1 (en) * 2006-11-16 2010-05-13 General Electric Company Sequential analysis of biological samples
US20100255606A1 (en) * 2007-10-05 2010-10-07 Nobuhiko Sato Labeled peptide having activity of binding to immunoglobulin and/or immunoglobulin complex and method of detecting or assaying immunoglobulin by using the peptide
US20100261203A1 (en) * 2009-04-07 2010-10-14 National Institute Of Transplantation Foundation Methods and kits for screening transplant recipients and candidates
US20100279307A1 (en) * 2003-12-18 2010-11-04 Merck Serono Sa Angiogenesis Inhibiting Molecules, Their Selection, Production and Their Use in the Treatment of Cancer
US20100330592A1 (en) * 2008-01-29 2010-12-30 Key Marc E Method for detecting truncated molecules
US20110065594A1 (en) * 2009-09-17 2011-03-17 Battelle Energy Alliance, Llc Identification of discriminant proteins through antibody profiling, methods and apparatus for identifying an individual
US20110065601A1 (en) * 2009-09-17 2011-03-17 Battelle Energy Alliance, Llc Identification of discriminant proteins through antibody profiling, methods and apparatus for identifying an individual
US20110092381A1 (en) * 2009-10-21 2011-04-21 General Electric Company Detection of plurality of targets in biological samples
US8323903B2 (en) 2001-10-12 2012-12-04 Life Technologies Corporation Antibody complexes and methods for immunolabeling
USRE44539E1 (en) 2001-05-10 2013-10-15 United States Department Of Energy Rapid classification of biological components
US20130323717A1 (en) * 2009-12-15 2013-12-05 MuHyeon CHOE Repeat-chain for the production of dimer, multimer, multimer complex and super-complex
US20140212889A1 (en) * 2011-09-09 2014-07-31 Tohoku University Method for staining tissue
US20150102234A1 (en) * 2012-05-09 2015-04-16 Life Technologies Corporation Systems and method for fluorescence imaging
WO2015184144A1 (en) * 2014-05-28 2015-12-03 Siemens Healthcare Diagnostics Inc. Rare molecule signal amplification
AT517379A1 (en) * 2015-06-26 2017-01-15 Technische Universität Graz fusion protein
USRE46351E1 (en) 2001-05-10 2017-03-28 Battelle Energy Alliance, Llc Antibody profiling sensitivity through increased reporter antibody layering
US9739773B1 (en) 2010-08-13 2017-08-22 David Gordon Bermudes Compositions and methods for determining successful immunization by one or more vaccines
US9944972B2 (en) 2014-03-11 2018-04-17 President And Fellows Of Harvard College High-throughput and highly multiplexed imaging with programmable nucleic acid probes
US10024796B2 (en) 2010-10-29 2018-07-17 President And Fellows Of Harvard College Nucleic acid nanostructure barcode probes
US20190000988A1 (en) * 2016-06-02 2019-01-03 Ultivue, Inc. Compositions and methods to expedite antibody-based exchange imaging
US10557851B2 (en) 2012-03-27 2020-02-11 Ventana Medical Systems, Inc. Signaling conjugates and methods of use
EP3734278A4 (en) * 2017-12-27 2020-12-30 Konica Minolta, Inc. Information acquisition system
WO2021084116A3 (en) * 2018-05-03 2021-06-10 Mursla Limited Biosensor activation and conditioning method
US11092606B2 (en) 2015-08-07 2021-08-17 President And Fellows Of Harvard College Super resolution imaging of protein-protein interactions
CN114002438A (en) * 2021-11-04 2022-02-01 河南赛诺特生物技术有限公司 Homogeneous immunohistochemical staining method for mouse-anti-mouse tissue
US11536715B2 (en) 2013-07-30 2022-12-27 President And Fellows Of Harvard College Quantitative DNA-based imaging and super-resolution imaging
CN116004221A (en) * 2022-12-15 2023-04-25 湖南卓润生物科技有限公司 New application of cyclic peptide, acridine labeled complex, preparation method and detection kit
US11754562B2 (en) 2016-12-09 2023-09-12 Ultivue, Inc. Methods for multiplex imaging using labeled nucleic acid imaging agents

Families Citing this family (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005050206A2 (en) 2003-09-17 2005-06-02 Molecular Probes, Inc. Competitive immunoassay
US20080003599A1 (en) * 2004-12-28 2008-01-03 Dary Ekaterina L Biological Microchip for Multiple Parallel Immunoassay of Compounds and Immunoassay Metods Using Said Microchip
BRPI0717416A2 (en) 2006-09-21 2013-11-12 Prometheus Lab Inc METHOD FOR PERFORMING A HIGH PRODUCTIVITY COMPLEX IMMUNOASON, AND
US20080213797A1 (en) * 2007-03-01 2008-09-04 Abbott Laboratories Immunoassays exhibiting a reduction in prozone phenomena
BRPI0813583A2 (en) * 2007-07-13 2014-12-30 Prometheus Lab Inc METHODS FOR SELECTING ANTICANUS MEDICINAL PRODUCT, IDENTIFYING A PULMONARY TUMOR RESPONSE, AND PROGNOSING A PATIENT'S RESPONSE, AND ARRANGEMENT
DK2250498T3 (en) 2008-02-25 2013-02-04 Nestec Sa PHARMACEUTICAL CHOICES FOR BREAST CANCER THERAPY USING ANTIBODY-BASED ARRAYS
JP5120889B2 (en) * 2008-06-12 2013-01-16 独立行政法人産業技術総合研究所 Boron dipyrromethane derivative and lipid peroxide measuring reagent using the same
WO2010009459A1 (en) * 2008-07-18 2010-01-21 Bayer Healthcare Llc Methods, devices, and systems for glycated hemoglobin analysis
ES2627909T3 (en) 2009-07-15 2017-08-01 Diatech Holdings, Inc. Drug selection for the treatment of gastric cancer using antibody-based matrices
CZ304250B6 (en) * 2010-08-27 2014-01-29 Ústav molekulární genetiky AV ČR, v.v.i. Set of mutually recognizable nanoparticles, process of their preparation as well as their use for multiple ultra-structural identification
WO2012103444A2 (en) * 2011-01-28 2012-08-02 Purdue Research Foundation Immunogenic compositions and reagents for preparing
US9719995B2 (en) 2011-02-03 2017-08-01 Pierian Holdings, Inc. Drug selection for colorectal cancer therapy using receptor tyrosine kinase profiling
US9044518B2 (en) 2011-03-30 2015-06-02 Arizona Board Of Regents, A Body Corporate Of The State Of Arizona Acting For And On Behalf Of Arizona State University Auristatin tyramine phosphate salts and auristatin aminoquinoline derivatives and prodrugs thereof
ES2553456T3 (en) 2011-09-02 2015-12-09 Nestec S.A. Profile path protein profiling to determine therapeutic efficacy
KR20150115733A (en) * 2012-12-11 2015-10-14 더 가버닝 카운슬 오브 더 유니버시티 오브 토론토 Wireless communication device-based detection system
EP3019559A4 (en) 2013-08-22 2017-04-05 Sony Corporation Water soluble fluorescent or colored dyes and methods for their use
JP6660392B2 (en) 2014-12-18 2020-03-18 ロード・アイランド・ホスピタルRhode Island Hospital Visualization of bacterial colonization and biofilm formation in orthopedic trauma explants
EP3262022A4 (en) 2015-02-26 2018-08-15 Sony Corporation Phenylethynylnaphthalene dyes and methods for their use
CN107454903B (en) 2015-02-26 2021-01-01 索尼公司 Water-soluble fluorescent or colored dyes containing conjugated groups
WO2016178083A2 (en) 2015-05-01 2016-11-10 Diagnostear, Ltd. Method for measuring tear constitutents in a tear sample
US10865310B2 (en) 2015-05-11 2020-12-15 Sony Corporation Of America Ultra bright dimeric or polymeric dyes
US10845305B1 (en) * 2015-11-20 2020-11-24 Verily Life Sciences Llc Method for increasing sensor resolution by spectrally stacking responsive dyes
CN109068971A (en) * 2016-01-14 2018-12-21 黛尔格诺斯蒂尔有限公司 The method for measuring the tear component in ocular fluid samples
CN105784657A (en) * 2016-03-25 2016-07-20 鲁延军 Method for efficiently screening positive clones of monoclonal antibody by using TR-FRET (Time-Resolved Fluorescence Resonance Energy Transfer)
EP3438666B1 (en) * 2016-03-31 2020-10-14 ABSOLOGY Co., Ltd Method for highly sensitive biomarker quantification using photo-oxidation amplification
CA3018564A1 (en) 2016-04-01 2017-10-05 Sony Corporation Ultra bright dimeric or polymeric fluorescent and colored dyes
US11434377B2 (en) 2016-04-01 2022-09-06 Sony Corporation Ultra bright dimeric or polymeric dyes with rigid spacing groups
AU2017248165B2 (en) 2016-04-06 2021-08-26 Sony Group Corporation Ultra bright dimeric or polymeric dyes with spacing linker groups
JP7068192B2 (en) 2016-05-10 2022-05-16 ソニーグループ株式会社 Compositions containing polymer dyes and cyclodextrins, and their use
JP2019519490A (en) 2016-05-10 2019-07-11 ソニー株式会社 Ultra bright polymer dye having a peptide backbone
CN109071961B (en) 2016-05-11 2021-04-27 索尼公司 Ultra-bright dimeric or polymeric dyes
GB201609235D0 (en) * 2016-05-25 2016-07-06 Univ Cape Town Production of a horseradish peroxidase-IGG fusion protein
US11377563B2 (en) 2016-06-06 2022-07-05 Sony Corporation Ionic polymers comprising fluorescent or colored reporter groups
US20200064341A1 (en) * 2017-03-14 2020-02-27 Nanotag Biotechnologies Gmbh Target detection using a monovalent antibody
EP3691690A1 (en) 2017-10-05 2020-08-12 Sony Corporation Programmable polymeric drugs
EP3710062A1 (en) 2017-11-16 2020-09-23 Sony Corporation Programmable polymeric drugs
KR102047978B1 (en) * 2017-11-22 2019-11-22 바디텍메드(주) Method for diagnosing rheumatoid arthritis based on lateral flow assay using anti-CCP antibody and Rheumatoid Factor
WO2019182765A1 (en) 2018-03-19 2019-09-26 Sony Corporation Use of divalent metals for enhancement of fluorescent signals
US20210032474A1 (en) 2018-03-21 2021-02-04 Sony Corporation Polymeric tandem dyes with linker groups
WO2021062176A2 (en) 2019-09-26 2021-04-01 Sony Corporation Polymeric tandem dyes with linker groups
WO2023044574A1 (en) * 2021-09-24 2023-03-30 Enveric Biosciences Canada Inc. Phosphorylated and sulfonated mescaline derivatives and methods of using

Citations (85)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5896A (en) * 1848-10-31 Shoe-pegging machine
US3996345A (en) * 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4174384A (en) * 1975-06-30 1979-11-13 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4196265A (en) * 1977-06-15 1980-04-01 The Wistar Institute Method of producing antibodies
US4199559A (en) * 1974-08-12 1980-04-22 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4235869A (en) * 1978-05-16 1980-11-25 Syva Company Assay employing a labeled Fab-fragment ligand complex
US4261968A (en) * 1979-05-10 1981-04-14 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4481298A (en) * 1981-04-13 1984-11-06 Amf Incorporated Pre-precipitated double antibody immunoassay method
US4603209A (en) * 1984-09-07 1986-07-29 The Regents Of The University Of California Fluorescent indicator dyes for calcium ions
US4603290A (en) * 1983-12-29 1986-07-29 Mitsubishi Denki Kabushiki Kaisha Constant-current generating circuit
US4642334A (en) * 1982-03-15 1987-02-10 Dnax Research Institute Of Molecular And Cellular Biology, Inc. Hybrid DNA prepared binding composition
US4661444A (en) * 1984-03-29 1987-04-28 Ortho Diagnostic Systems Inc. Homogeneous immunoassays employing double antibody conjugates comprising anti-idiotype antibody
US4714763A (en) * 1985-07-11 1987-12-22 Viomedics Inc. Novel oxazine-ureas and thiazine urea chromophors as fluorescent labels
US4735210A (en) * 1985-07-05 1988-04-05 Immunomedics, Inc. Lymphographic and organ imaging method and kit
US4737579A (en) * 1984-11-02 1988-04-12 Oncogen Monoclonal antibodies for human non-small cell lung carinomas
US4774339A (en) * 1987-08-10 1988-09-27 Molecular Probes, Inc. Chemically reactive dipyrrometheneboron difluoride dyes
US4810636A (en) * 1986-12-09 1989-03-07 Miles Inc. Chromogenic acridinone enzyme substrates
US4812409A (en) * 1986-01-31 1989-03-14 Eastman Kodak Company Hydrolyzable fluorescent substrates and analytical determinations using same
US4849362A (en) * 1988-05-19 1989-07-18 Smithkline Beckman Corporation Fluorescent intracellular calcium indicators
US4945171A (en) * 1987-08-10 1990-07-31 Molecular Probes, Inc. Xanthene dyes having a fused (C) benzo ring
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US4981977A (en) * 1989-06-09 1991-01-01 Carnegie-Mellon University Intermediate for and fluorescent cyanine dyes containing carboxylic acid groups
US5011771A (en) * 1984-04-12 1991-04-30 The General Hospital Corporation Multiepitopic immunometric assay
US5034506A (en) * 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US5082928A (en) * 1985-06-17 1992-01-21 Best Mark P Method of preparing conjugated antibodies
US5084398A (en) * 1987-11-20 1992-01-28 Creative Biomolecules Selective removal of immune complexes
US5132432A (en) * 1989-09-22 1992-07-21 Molecular Probes, Inc. Chemically reactive pyrenyloxy sulfonic acid dyes
US5151507A (en) * 1986-07-02 1992-09-29 E. I. Du Pont De Nemours And Company Alkynylamino-nucleotides
US5187085A (en) * 1990-09-28 1993-02-16 Applied Biosystems, Inc. Nucleic acid sequence analysis with nucleoside-5'-o-(1-thiotriphosphates)
US5187288A (en) * 1991-05-22 1993-02-16 Molecular Probes, Inc. Ethenyl-substituted dipyrrometheneboron difluoride dyes and their synthesis
US5227487A (en) * 1990-04-16 1993-07-13 Molecular Probes, Inc. Certain tricyclic and pentacyclic-hetero nitrogen rhodol dyes
US5229302A (en) * 1988-03-29 1993-07-20 Matsushita Electric Industrial Co., Ltd. Fluorescence immunoassay method utilizing pseudo-antigens combined with fluorescent quenchers
US5248782A (en) * 1990-12-18 1993-09-28 Molecular Probes, Inc. Long wavelength heteroaryl-substituted dipyrrometheneboron difluoride dyes
US5268486A (en) * 1986-04-18 1993-12-07 Carnegie-Mellon Unversity Method for labeling and detecting materials employing arylsulfonate cyanine dyes
US5274113A (en) * 1991-11-01 1993-12-28 Molecular Probes, Inc. Long wavelength chemically reactive dipyrrometheneboron difluoride dyes and conjugates
US5360895A (en) * 1987-04-22 1994-11-01 Associated Universities, Inc. Derivatized gold clusters and antibody-gold cluster conjugates
US5443953A (en) * 1993-12-08 1995-08-22 Immunomedics, Inc. Preparation and use of immunoconjugates
US5451343A (en) * 1991-05-20 1995-09-19 Spectra Group Limited, Inc. Fluorone and pyronin y derivatives
US5459276A (en) * 1994-05-20 1995-10-17 Molecular Probes, Inc. Benzazolylcoumarin-based ion indicators for heavy metals
US5501980A (en) * 1994-05-20 1996-03-26 Molecular Probes, Inc. Benzazolylcoumarin-based ion indicators
US5558991A (en) * 1986-07-02 1996-09-24 E. I. Du Pont De Nemours And Company DNA sequencing method using acyclonucleoside triphosphates
US5565332A (en) * 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5569587A (en) * 1986-04-18 1996-10-29 Carnegie Mellon University Method for labeling and detecting materials employing luminescent arysulfonate cyanine dyes
US5585243A (en) * 1993-09-15 1996-12-17 The Blood Center Of Southeastern Wisconsin, Inc. Method of detecting cytopenia that is mediated by drug-dependent antibody binding to blood cells
US5602240A (en) * 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5627027A (en) * 1986-04-18 1997-05-06 Carnegie Mellon University Cyanine dyes as labeling reagents for detection of biological and other materials by luminescence methods
US5630924A (en) * 1995-04-20 1997-05-20 Perseptive Biosystems, Inc. Compositions, methods and apparatus for ultrafast electroseparation analysis
US5637258A (en) * 1996-03-18 1997-06-10 Nanocrystals Technology L.P. Method for producing rare earth activited metal oxide nanocrystals
US5679519A (en) * 1995-05-09 1997-10-21 Oprandy; John J. Multi-label complex for enhanced sensitivity in electrochemiluminescence assay
US5736137A (en) * 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5789157A (en) * 1990-06-11 1998-08-04 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: tissue selex
US5798276A (en) * 1995-06-07 1998-08-25 Molecular Probes, Inc. Reactive derivatives of sulforhodamine 101 with enhanced hydrolytic stability
US5808044A (en) * 1993-01-22 1998-09-15 Pharmacia Biotech Inc. Indocarbocyanine and benzindocarbocyanine phosphoramidites
US5846737A (en) * 1996-07-26 1998-12-08 Molecular Probes, Inc. Conjugates of sulforhodamine fluorophores with enhanced fluorescence
US5872221A (en) * 1986-07-18 1999-02-16 The University Of Melbourne Antibody having binding specificity to parathyroid hormone related protein (PTHrP) and kit comprising the same
US5877310A (en) * 1997-04-25 1999-03-02 Carnegie Mellon University Glycoconjugated fluorescent labeling reagents
US5948386A (en) * 1997-03-14 1999-09-07 The Curators Of The University Of Missouri Conjugate and method for forming aminomethyl phosphorus conjugates
US5969135A (en) * 1995-11-02 1999-10-19 Icn Pharmaceuticals, Inc. Oligonucleotide analogs with an amino acid or a modified amino alcohol residue
US6002003A (en) * 1998-04-14 1999-12-14 Beckman Instruments, Inc. Cyanine dye activating group with improved coupling selectivity
US6004536A (en) * 1995-11-14 1999-12-21 Molecular Probes, Inc. Lipophilic cyanine dyes with enchanced aqueous solubilty
US6008373A (en) * 1995-06-07 1999-12-28 Carnegie Mellon University Fluorescent labeling complexes with large stokes shift formed by coupling together cyanine and other fluorochromes capable of resonance energy transfer
US6043025A (en) * 1995-04-20 2000-03-28 Carnegie Mellon University Difference gel electrophoresis using matched multiple dyes
US6080868A (en) * 1998-01-23 2000-06-27 The Perkin-Elmer Corporation Nitro-substituted non-fluorescent asymmetric cyanine dye compounds
US6114038A (en) * 1998-11-10 2000-09-05 Biocrystal Ltd. Functionalized nanocrystals and their use in detection systems
US6133445A (en) * 1997-12-17 2000-10-17 Carnegie Mellon University Rigidized trimethine cyanine dyes
US6166202A (en) * 1996-02-05 2000-12-26 Amersham Pharmacia Biotech Uk Limited Benzophenoxazine dyes
US6179912B1 (en) * 1999-12-20 2001-01-30 Biocrystal Ltd. Continuous flow process for production of semiconductor nanocrystals
US6214568B1 (en) * 1991-02-14 2001-04-10 Roche Diagnostics Gmbh Monoclonal antibodies of isotype IgG which bind specifically to glutamate decarboxylase, and uses thereof
US6221602B1 (en) * 1998-11-10 2001-04-24 Bio-Pixels Ltd. Functionalized nanocrystals and their use in labeling for strand synthesis or sequence determination
US6229055B1 (en) * 1996-04-12 2001-05-08 Molecular Probes, Inc. Synthesis of fluorinated xanthene derivatives
US6261779B1 (en) * 1998-11-10 2001-07-17 Bio-Pixels Ltd. Nanocrystals having polynucleotide strands and their use to form dendrimers in a signal amplification system
US6306610B1 (en) * 1998-09-18 2001-10-23 Massachusetts Institute Of Technology Biological applications of quantum dots
US6326144B1 (en) * 1998-09-18 2001-12-04 Massachusetts Institute Of Technology Biological applications of quantum dots
US6339392B1 (en) * 1999-01-13 2002-01-15 Honda Giken Kogyo Kabushiki Kaisha Gate for radar-mounted vehicle, having partition walls not grouped or detected by radar apparatus
US6345596B1 (en) * 1999-04-07 2002-02-12 Ina Walzlager Schaeffler Ohg Engageable cam follower or engageable lifter element
US6399392B1 (en) * 1999-04-23 2002-06-04 Molecular Probes, Inc. Xanthene dyes and their application as luminescence quenching compounds
US20020077487A1 (en) * 2000-09-29 2002-06-20 Molecular Probes, Inc. Modified carbocyanine dyes and their conjugates
US20020132254A1 (en) * 2000-11-30 2002-09-19 Twu Jesse J. Molecular labeling and assay systems using poly (amino acid)-metal ion complexes as linkers
US6475808B1 (en) * 1998-07-14 2002-11-05 Zyomyx, Incorporated Arrays of proteins and methods of use thereof
US6482655B1 (en) * 1993-07-23 2002-11-19 University Of Utah Research Foundation Immunoassay procedure utilizing fluorogenic tracer antigens
US6787638B1 (en) * 1998-12-02 2004-09-07 Applied Molecular Evolution, Inc. Tumor specific human monoclonal antibodies and methods of use
US6855551B2 (en) * 1998-09-18 2005-02-15 Massachusetts Institute Of Technology Biological applications of quantum dots
US6972326B2 (en) * 2001-12-03 2005-12-06 Molecular Probes, Inc. Labeling of immobilized proteins using dipyrrometheneboron difluoride dyes
US20070269902A1 (en) * 2001-10-12 2007-11-22 Joseph Beechem Antibody complexes and methods for immunolabeling
US20090124511A1 (en) * 2001-10-12 2009-05-14 Invitrogen Corporation Antibody complexes and methods for immunolabeling

Family Cites Families (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6033479B2 (en) 1980-07-30 1985-08-02 協和醗酵工業株式会社 Method for quantifying hydrogen peroxide
US5055556A (en) 1981-10-06 1991-10-08 The Board Of Trustees Of The Leland Stanford Jr. Univ. Fluorescent conjugates for analysis of molecules and cells
US4859582A (en) 1981-10-06 1989-08-22 The Board Of Trustees Of The Leland Stanford Jr. University Fluorescent conjugates for analysis of molecules and cells
US4520110A (en) 1981-10-06 1985-05-28 The Board Of Trustees Of The Leland Stanford Junior University Fluorescent immunoassay employing a phycobiliprotein labeled ligand or receptor
US4469787A (en) 1982-05-14 1984-09-04 Mallinckrodt Inc. Immunoassay involving soluble complex of second antibody and labeled binding protein
US4659678A (en) 1982-09-29 1987-04-21 Serono Diagnostics Limited Immunoassay of antigens
US4542104A (en) 1983-04-06 1985-09-17 The Board Of Trustees Of The Leland Stanford Jr. Univ. Phycobiliprotein fluorescent conjugates
JPS6082966A (en) * 1983-10-14 1985-05-11 Amano Pharmaceut Co Ltd Assay of antigen
EP0150905A3 (en) 1984-01-05 1987-01-14 Ortho Diagnostic Systems Inc. Anti-idiotype assay using fluorescent energy transfer
US4665024A (en) 1984-10-01 1987-05-12 Becton, Dickinson And Company Fluorescent gram stain
US4659839A (en) 1984-10-10 1987-04-21 Mallinckrodt, Inc. Coupling agents for radiolabeled antibody fragments
US5422266A (en) 1984-12-31 1995-06-06 University Of Georgia Research Foundation, Inc. Recombinant DNA vectors capable of expressing apoaequorin
NL8501219A (en) 1985-04-29 1986-11-17 Stichting Vrienden Van De Stic IMMUNOLOGICAL COMPLEX, ITS PREPARATION AND APPLICATION.
US4959306A (en) 1986-11-28 1990-09-25 Sclavo, Inc. Labeling design for a binding assay reagent
DE3800048A1 (en) 1987-05-14 1988-11-24 Boehringer Mannheim Gmbh METHOD FOR DETERMINING AN ANTIBODY IN HUMAN BODY LIQUIDS
DE3717401A1 (en) 1987-05-23 1988-12-08 Behringwerke Ag One-step immunoassay for the determination of antigen-specific antibodies from one of the immunoglobulin classes A, M, D or E and a suitable agent
US4814433A (en) 1987-09-16 1989-03-21 Miles Inc. Method for obtaining a papain-free antibody fragment preparation
NO884368L (en) 1987-10-02 1989-04-03 Du Pont IMMUNAL ANALYSIS USING IGG CITIZING ANTIBODY AND MULTIPLE, MONOCLONAL ANTIBODY DISEASE SYSTEM.
JPH01131458A (en) * 1987-11-17 1989-05-24 Nippon Zenyaku Kogyo Kk Reagent for simple and rapid immunoassay
US4891313A (en) 1988-01-21 1990-01-02 Boehringer Manheim Corporation Method for determination of a component of a sample
JPH0276600A (en) * 1988-09-10 1990-03-15 Akad Wissenschaften Ddr Molecular probe with a mark, its production and use
DE3836348A1 (en) 1988-10-25 1990-04-26 Boehringer Mannheim Gmbh METHOD FOR DETERMINING AN ANTIQUE CLASS-SPECIFIC ANTIBODY AND REAGENT SUITABLE FOR IT
EP0368674A3 (en) 1988-11-11 1991-10-09 SANYO CHEMICAL INDUSTRIES, Ltd. Immunoassay and test kits therefor
CA2013006A1 (en) * 1989-03-24 1990-09-24 Marc E. Key Reagent complex for immunoassay
US5196306A (en) 1989-03-29 1993-03-23 E. I. Du Pont De Nemours And Company Method for the detection or quantitation of an analyte using an analyte dependent enzyme activation system
GB8910263D0 (en) 1989-05-04 1989-06-21 Ciba Geigy Ag Monoclonal antibodies specific for an immunoglobulin isotype
US5089419A (en) * 1989-08-07 1992-02-18 International Canine Genetics Detection of pregnancy by identification of the c peptide of relaxin in the urine of animals
EP0489061A4 (en) * 1989-08-24 1992-10-14 Australian Nuclear Science And Technology Organisation Radio-labelled antibodies for imaging
US6274324B1 (en) 1989-12-01 2001-08-14 Unilever Patent Holdings B.V. Specific binding reagent comprising a variable domain protein linked to a support or tracer
US5292658A (en) 1989-12-29 1994-03-08 University Of Georgia Research Foundation, Inc. Boyd Graduate Studies Research Center Cloning and expressions of Renilla luciferase
US5433896A (en) 1994-05-20 1995-07-18 Molecular Probes, Inc. Dibenzopyrrometheneboron difluoride dyes
US5208148A (en) 1990-12-07 1993-05-04 Molecular Probes, Inc. Lipophilic fluorescent glycosidase substrates
GR920100123A (en) * 1991-04-12 1993-03-16 Ortho Diagnostic Systems Inc Multiple color staining reagent and method of analysis.
US6352830B1 (en) 1991-08-22 2002-03-05 The Board Of Trustees Of The Leland Stanford Junior University NF-AT polypeptides and polynucleotides and screening methods for immunosuppressive agents
US5242805A (en) 1991-08-23 1993-09-07 Molecular Probes, Inc. Long wavelength lipophilic fluorogenic glycosidase substrates
EP0603266B1 (en) 1991-08-23 1999-05-12 Molecular Probes, Inc. Use of haloalkyl derivatives of reporter molecules to analyze metabolic activity in cells
US5576424A (en) 1991-08-23 1996-11-19 Molecular Probes, Inc. Haloalkyl derivatives of reporter molecules used to analyze metabolic activity in cells
US5316906A (en) 1991-08-23 1994-05-31 Molecular Probes, Inc. Enzymatic analysis using substrates that yield fluorescent precipitates
CA2093494A1 (en) 1992-04-17 1993-10-18 Keisuke Iwata Method for the elimination of non-specific reactions in immuno-assays
US5195303A (en) 1992-05-29 1993-03-23 Philip Morris Incorporated Apparatus for automatic bagging of compressed tobacco
US6030773A (en) 1992-07-08 2000-02-29 Agnello; Vincent Chemiluminescent assay for DSDNA antibodies
US5281521A (en) 1992-07-20 1994-01-25 The Trustees Of The University Of Pennsylvania Modified avidin-biotin technique
DE59309999D1 (en) 1992-11-23 2000-05-11 Sanochemia Pharmazeutika Ag Wi Methods for the detection of antibodies and antigens
ES2051651B1 (en) * 1992-12-10 1995-01-01 Univ Salamanca PROCEDURE FOR THE SIMULTANEOUS QUANTIFICATION, IN A SINGLE MEASUREMENT, OF THE MAIN TYPES OF HUMAN LYMPHOCYTES AND THEIR SUB-POPULATIONS.
EP0629857A3 (en) 1993-06-16 1996-06-26 Nisshin Flour Milling Co Immunoassay for quantitatively determining antigens.
US5561045A (en) 1994-01-04 1996-10-01 Intracel Corporation Detection reagent, article, and immunoassay method
US5869274A (en) 1995-08-09 1999-02-09 Zymed Laboratories, Inc. Immuno-histochemical method that reduces background staining
US5981180A (en) 1995-10-11 1999-11-09 Luminex Corporation Multiplexed analysis of clinical specimens apparatus and methods
US5736330A (en) 1995-10-11 1998-04-07 Luminex Corporation Method and compositions for flow cytometric determination of DNA sequences
WO1997017610A1 (en) * 1995-11-08 1997-05-15 Pharmaceutical Discovery Corporation Method for fluorescent labeling of antibody
US5994143A (en) * 1996-02-01 1999-11-30 Abbott Laboratories Polymeric fluorophores enhanced by moieties providing a hydrophobic and conformationally restrictive microenvironment
US5723294A (en) 1996-03-05 1998-03-03 Gull Laboratories Methods for detection and discrimination of multiple analytes using fluorescent technology
WO1997040104A1 (en) 1996-04-19 1997-10-30 Amersham Pharmacia Biotech Uk Limited Squarate dyes and their use in fluorescent sequencing method
GB9712818D0 (en) 1996-07-08 1997-08-20 Cambridge Antibody Tech Labelling and selection of specific binding molecules
US5922845A (en) 1996-07-11 1999-07-13 Medarex, Inc. Therapeutic multispecific compounds comprised of anti-Fcα receptor antibodies
US5696157A (en) 1996-11-15 1997-12-09 Molecular Probes, Inc. Sulfonated derivatives of 7-aminocoumarin
US5830912A (en) 1996-11-15 1998-11-03 Molecular Probes, Inc. Derivatives of 6,8-difluoro-7-hydroxycoumarin
ZA9710262B (en) 1996-11-19 1999-05-13 Lilly Co Eli Process for the synthesis of benzothiophenes
US5945294A (en) 1996-11-26 1999-08-31 Heska Corporation Method to detect IgE
US5773236A (en) 1997-04-25 1998-06-30 Molecule Probes, Inc. Assay for glutathiane transferase using polyhaloaryl-substituted reporter molecules
US5994089A (en) * 1997-05-16 1999-11-30 Coulter International Corp. Simultaneous analyses of white blood cell subsets using multi-color, multi-intensity fluorescent markers in flow cytometry
US5891741A (en) 1997-05-16 1999-04-06 Coulter International Corp. Antibody-aminodextran-phycobiliprotein conjugates
US6146836A (en) * 1997-05-19 2000-11-14 Bayer Corporation Immunoassays using anti-allotypic monoclonal antibodies
US6130101A (en) 1997-09-23 2000-10-10 Molecular Probes, Inc. Sulfonated xanthene derivatives
JPH11153599A (en) 1997-11-21 1999-06-08 Dai Ichi Pure Chem Co Ltd Immunoassay
AU3386399A (en) 1998-04-08 1999-10-25 Ewald A. Terpetschnig Luminescent compounds
CN1163753C (en) 1998-04-14 2004-08-25 大塚制药株式会社 Method for assaying antibody and device for assaying antibody
US6150123A (en) 1998-09-03 2000-11-21 Centre Integre De Recherches Biocliniques Sur Le Sida (Cirbs) Affinity biotinylation
JP2000214165A (en) 1999-01-22 2000-08-04 Fuji Photo Film Co Ltd Homogeneous system enzyme immunity analyzing method
JP2000310638A (en) 1999-04-28 2000-11-07 Fuji Photo Film Co Ltd Uniform system enzyme immunity analysis method
US6664047B1 (en) 1999-04-30 2003-12-16 Molecular Probes, Inc. Aza-benzazolium containing cyanine dyes
DE69941067D1 (en) 1999-07-02 2009-08-13 Visen Medical Inc Fluorescent cyanine labels with a sulphamido linker
JP2001059845A (en) 1999-08-24 2001-03-06 Fuji Photo Film Co Ltd Method and element for dry analysis
AU7316900A (en) 1999-09-20 2001-04-24 Fuji Photo Film Co., Ltd. Compounds for fluorescence labeling
US20020081635A1 (en) 2000-05-11 2002-06-27 Thomas Terry E. Novel antibody compositions for preparing enriched T cell preparations
US7524628B2 (en) 2000-07-25 2009-04-28 The Trustees Of The University Of Pennsylvania Method for detecting molecules expressing a selected epitope via fluorescent dyes
ATE415399T1 (en) 2000-08-04 2008-12-15 Molecular Probes Inc CONDENSED RINGS CONTAINING 1,2-DIHYDRO-7-HYDROXYCHINOLINE DERIVATIVES

Patent Citations (94)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5896A (en) * 1848-10-31 Shoe-pegging machine
US3996345A (en) * 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4199559A (en) * 1974-08-12 1980-04-22 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4174384A (en) * 1975-06-30 1979-11-13 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4196265A (en) * 1977-06-15 1980-04-01 The Wistar Institute Method of producing antibodies
US4235869A (en) * 1978-05-16 1980-11-25 Syva Company Assay employing a labeled Fab-fragment ligand complex
US4261968A (en) * 1979-05-10 1981-04-14 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4481298A (en) * 1981-04-13 1984-11-06 Amf Incorporated Pre-precipitated double antibody immunoassay method
US4642334A (en) * 1982-03-15 1987-02-10 Dnax Research Institute Of Molecular And Cellular Biology, Inc. Hybrid DNA prepared binding composition
US4603290A (en) * 1983-12-29 1986-07-29 Mitsubishi Denki Kabushiki Kaisha Constant-current generating circuit
US4661444A (en) * 1984-03-29 1987-04-28 Ortho Diagnostic Systems Inc. Homogeneous immunoassays employing double antibody conjugates comprising anti-idiotype antibody
US5011771A (en) * 1984-04-12 1991-04-30 The General Hospital Corporation Multiepitopic immunometric assay
US4603209A (en) * 1984-09-07 1986-07-29 The Regents Of The University Of California Fluorescent indicator dyes for calcium ions
US4737579A (en) * 1984-11-02 1988-04-12 Oncogen Monoclonal antibodies for human non-small cell lung carinomas
US5034506A (en) * 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US5082928A (en) * 1985-06-17 1992-01-21 Best Mark P Method of preparing conjugated antibodies
US4735210A (en) * 1985-07-05 1988-04-05 Immunomedics, Inc. Lymphographic and organ imaging method and kit
US4714763A (en) * 1985-07-11 1987-12-22 Viomedics Inc. Novel oxazine-ureas and thiazine urea chromophors as fluorescent labels
US4812409A (en) * 1986-01-31 1989-03-14 Eastman Kodak Company Hydrolyzable fluorescent substrates and analytical determinations using same
US5268486A (en) * 1986-04-18 1993-12-07 Carnegie-Mellon Unversity Method for labeling and detecting materials employing arylsulfonate cyanine dyes
US5627027A (en) * 1986-04-18 1997-05-06 Carnegie Mellon University Cyanine dyes as labeling reagents for detection of biological and other materials by luminescence methods
US5569766A (en) * 1986-04-18 1996-10-29 Carnegie Mellon University Method for labeling and detecting materials employing arylsulfonate cyanine dyes
US5486616A (en) * 1986-04-18 1996-01-23 Carnegie Mellon University Method for labeling and detecting materials employing arylsulfonate cyanine dyes
US5569587A (en) * 1986-04-18 1996-10-29 Carnegie Mellon University Method for labeling and detecting materials employing luminescent arysulfonate cyanine dyes
US5558991A (en) * 1986-07-02 1996-09-24 E. I. Du Pont De Nemours And Company DNA sequencing method using acyclonucleoside triphosphates
US5151507A (en) * 1986-07-02 1992-09-29 E. I. Du Pont De Nemours And Company Alkynylamino-nucleotides
US5872221A (en) * 1986-07-18 1999-02-16 The University Of Melbourne Antibody having binding specificity to parathyroid hormone related protein (PTHrP) and kit comprising the same
US4810636A (en) * 1986-12-09 1989-03-07 Miles Inc. Chromogenic acridinone enzyme substrates
US5360895A (en) * 1987-04-22 1994-11-01 Associated Universities, Inc. Derivatized gold clusters and antibody-gold cluster conjugates
US4945171A (en) * 1987-08-10 1990-07-31 Molecular Probes, Inc. Xanthene dyes having a fused (C) benzo ring
US4774339A (en) * 1987-08-10 1988-09-27 Molecular Probes, Inc. Chemically reactive dipyrrometheneboron difluoride dyes
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5084398A (en) * 1987-11-20 1992-01-28 Creative Biomolecules Selective removal of immune complexes
US5229302A (en) * 1988-03-29 1993-07-20 Matsushita Electric Industrial Co., Ltd. Fluorescence immunoassay method utilizing pseudo-antigens combined with fluorescent quenchers
US4849362A (en) * 1988-05-19 1989-07-18 Smithkline Beckman Corporation Fluorescent intracellular calcium indicators
US4981977A (en) * 1989-06-09 1991-01-01 Carnegie-Mellon University Intermediate for and fluorescent cyanine dyes containing carboxylic acid groups
US5132432A (en) * 1989-09-22 1992-07-21 Molecular Probes, Inc. Chemically reactive pyrenyloxy sulfonic acid dyes
US5227487A (en) * 1990-04-16 1993-07-13 Molecular Probes, Inc. Certain tricyclic and pentacyclic-hetero nitrogen rhodol dyes
US5442045A (en) * 1990-04-16 1995-08-15 Molecular Probes, Inc. Biological conjugates of fluorescent rhodol dyes
US5789157A (en) * 1990-06-11 1998-08-04 Nexstar Pharmaceuticals, Inc. Systematic evolution of ligands by exponential enrichment: tissue selex
US5602240A (en) * 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5187085A (en) * 1990-09-28 1993-02-16 Applied Biosystems, Inc. Nucleic acid sequence analysis with nucleoside-5'-o-(1-thiotriphosphates)
US5248782A (en) * 1990-12-18 1993-09-28 Molecular Probes, Inc. Long wavelength heteroaryl-substituted dipyrrometheneboron difluoride dyes
US6214568B1 (en) * 1991-02-14 2001-04-10 Roche Diagnostics Gmbh Monoclonal antibodies of isotype IgG which bind specifically to glutamate decarboxylase, and uses thereof
US5451343A (en) * 1991-05-20 1995-09-19 Spectra Group Limited, Inc. Fluorone and pyronin y derivatives
US5187288A (en) * 1991-05-22 1993-02-16 Molecular Probes, Inc. Ethenyl-substituted dipyrrometheneboron difluoride dyes and their synthesis
US5565332A (en) * 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5274113A (en) * 1991-11-01 1993-12-28 Molecular Probes, Inc. Long wavelength chemically reactive dipyrrometheneboron difluoride dyes and conjugates
US5736137A (en) * 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5808044A (en) * 1993-01-22 1998-09-15 Pharmacia Biotech Inc. Indocarbocyanine and benzindocarbocyanine phosphoramidites
US6482655B1 (en) * 1993-07-23 2002-11-19 University Of Utah Research Foundation Immunoassay procedure utilizing fluorogenic tracer antigens
US5585243A (en) * 1993-09-15 1996-12-17 The Blood Center Of Southeastern Wisconsin, Inc. Method of detecting cytopenia that is mediated by drug-dependent antibody binding to blood cells
US5443953A (en) * 1993-12-08 1995-08-22 Immunomedics, Inc. Preparation and use of immunoconjugates
US5635603A (en) * 1993-12-08 1997-06-03 Immunomedics, Inc. Preparation and use of immunoconjugates
US5501980A (en) * 1994-05-20 1996-03-26 Molecular Probes, Inc. Benzazolylcoumarin-based ion indicators
US5459276A (en) * 1994-05-20 1995-10-17 Molecular Probes, Inc. Benzazolylcoumarin-based ion indicators for heavy metals
US5630924A (en) * 1995-04-20 1997-05-20 Perseptive Biosystems, Inc. Compositions, methods and apparatus for ultrafast electroseparation analysis
US6127134A (en) * 1995-04-20 2000-10-03 Carnegie Mellon University Difference gel electrophoresis using matched multiple dyes
US6043025A (en) * 1995-04-20 2000-03-28 Carnegie Mellon University Difference gel electrophoresis using matched multiple dyes
US5679519A (en) * 1995-05-09 1997-10-21 Oprandy; John J. Multi-label complex for enhanced sensitivity in electrochemiluminescence assay
US5798276A (en) * 1995-06-07 1998-08-25 Molecular Probes, Inc. Reactive derivatives of sulforhodamine 101 with enhanced hydrolytic stability
US6130094A (en) * 1995-06-07 2000-10-10 Carnegie Mellon University Reagents including a carrier and fluorescent labeling complexes with large stokes shift formed by coupling together cyanine and other fluorochromes capable of resonance energy transfer
US6008373A (en) * 1995-06-07 1999-12-28 Carnegie Mellon University Fluorescent labeling complexes with large stokes shift formed by coupling together cyanine and other fluorochromes capable of resonance energy transfer
US5969135A (en) * 1995-11-02 1999-10-19 Icn Pharmaceuticals, Inc. Oligonucleotide analogs with an amino acid or a modified amino alcohol residue
US6004536A (en) * 1995-11-14 1999-12-21 Molecular Probes, Inc. Lipophilic cyanine dyes with enchanced aqueous solubilty
US6166202A (en) * 1996-02-05 2000-12-26 Amersham Pharmacia Biotech Uk Limited Benzophenoxazine dyes
US5637258A (en) * 1996-03-18 1997-06-10 Nanocrystals Technology L.P. Method for producing rare earth activited metal oxide nanocrystals
US6229055B1 (en) * 1996-04-12 2001-05-08 Molecular Probes, Inc. Synthesis of fluorinated xanthene derivatives
US5846737A (en) * 1996-07-26 1998-12-08 Molecular Probes, Inc. Conjugates of sulforhodamine fluorophores with enhanced fluorescence
US5948386A (en) * 1997-03-14 1999-09-07 The Curators Of The University Of Missouri Conjugate and method for forming aminomethyl phosphorus conjugates
US5877310A (en) * 1997-04-25 1999-03-02 Carnegie Mellon University Glycoconjugated fluorescent labeling reagents
US6133445A (en) * 1997-12-17 2000-10-17 Carnegie Mellon University Rigidized trimethine cyanine dyes
US6080868A (en) * 1998-01-23 2000-06-27 The Perkin-Elmer Corporation Nitro-substituted non-fluorescent asymmetric cyanine dye compounds
US6541618B1 (en) * 1998-01-23 2003-04-01 Applera Corporation Nitro-substituted non-fluorescent asymmetric cyanine dye compounds
US6002003A (en) * 1998-04-14 1999-12-14 Beckman Instruments, Inc. Cyanine dye activating group with improved coupling selectivity
US6475808B1 (en) * 1998-07-14 2002-11-05 Zyomyx, Incorporated Arrays of proteins and methods of use thereof
US6855551B2 (en) * 1998-09-18 2005-02-15 Massachusetts Institute Of Technology Biological applications of quantum dots
US6326144B1 (en) * 1998-09-18 2001-12-04 Massachusetts Institute Of Technology Biological applications of quantum dots
US6306610B1 (en) * 1998-09-18 2001-10-23 Massachusetts Institute Of Technology Biological applications of quantum dots
US6114038A (en) * 1998-11-10 2000-09-05 Biocrystal Ltd. Functionalized nanocrystals and their use in detection systems
US6221602B1 (en) * 1998-11-10 2001-04-24 Bio-Pixels Ltd. Functionalized nanocrystals and their use in labeling for strand synthesis or sequence determination
US6261779B1 (en) * 1998-11-10 2001-07-17 Bio-Pixels Ltd. Nanocrystals having polynucleotide strands and their use to form dendrimers in a signal amplification system
US6787638B1 (en) * 1998-12-02 2004-09-07 Applied Molecular Evolution, Inc. Tumor specific human monoclonal antibodies and methods of use
US6339392B1 (en) * 1999-01-13 2002-01-15 Honda Giken Kogyo Kabushiki Kaisha Gate for radar-mounted vehicle, having partition walls not grouped or detected by radar apparatus
US6345596B1 (en) * 1999-04-07 2002-02-12 Ina Walzlager Schaeffler Ohg Engageable cam follower or engageable lifter element
US6399392B1 (en) * 1999-04-23 2002-06-04 Molecular Probes, Inc. Xanthene dyes and their application as luminescence quenching compounds
US6179912B1 (en) * 1999-12-20 2001-01-30 Biocrystal Ltd. Continuous flow process for production of semiconductor nanocrystals
US20020077487A1 (en) * 2000-09-29 2002-06-20 Molecular Probes, Inc. Modified carbocyanine dyes and their conjugates
US6974873B2 (en) * 2000-09-29 2005-12-13 Molecular Probes, Inc. Modified carbocyanine dyes and their conjugates
US6977305B2 (en) * 2000-09-29 2005-12-20 Molecular Probes, Inc. Modified carbocyanine dyes and their conjugates
US20020132254A1 (en) * 2000-11-30 2002-09-19 Twu Jesse J. Molecular labeling and assay systems using poly (amino acid)-metal ion complexes as linkers
US20070269902A1 (en) * 2001-10-12 2007-11-22 Joseph Beechem Antibody complexes and methods for immunolabeling
US20090124511A1 (en) * 2001-10-12 2009-05-14 Invitrogen Corporation Antibody complexes and methods for immunolabeling
US6972326B2 (en) * 2001-12-03 2005-12-06 Molecular Probes, Inc. Labeling of immobilized proteins using dipyrrometheneboron difluoride dyes

Cited By (101)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE44539E1 (en) 2001-05-10 2013-10-15 United States Department Of Energy Rapid classification of biological components
USRE46351E1 (en) 2001-05-10 2017-03-28 Battelle Energy Alliance, Llc Antibody profiling sensitivity through increased reporter antibody layering
US8535894B2 (en) 2001-10-12 2013-09-17 Life Technologies Corporation Antibody complexes and methods for immunolabeling
US8323903B2 (en) 2001-10-12 2012-12-04 Life Technologies Corporation Antibody complexes and methods for immunolabeling
US20050069962A1 (en) * 2001-10-12 2005-03-31 Archer Robert M Antibody complexes and methods for immunolabeling
US8093010B2 (en) * 2003-12-18 2012-01-10 Merck Serono S.A. Angiogenesis inhibiting molecules, their selection, production and their use in the treatment of cancer
US20100279307A1 (en) * 2003-12-18 2010-11-04 Merck Serono Sa Angiogenesis Inhibiting Molecules, Their Selection, Production and Their Use in the Treatment of Cancer
US20100041076A1 (en) * 2004-03-25 2010-02-18 Ez Bio Inc. Reagents, Methods and Kits for the Universal Rapid Immuno-Detection
US20050214882A1 (en) * 2004-03-25 2005-09-29 Ez Bio Inc. Reagents, methods and kits for the universal rapid immuno-detection
US7928193B2 (en) * 2004-07-08 2011-04-19 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Antigen binding proteins directed against scavenger receptor B1 that inhibit HCV replication
US20080286275A1 (en) * 2004-07-08 2008-11-20 Riccardo Cortese Antigen Binding Proteins Directed Against Scavenger Receptor B1 that Inhibit Hcv Replication
US20100179073A1 (en) * 2004-11-26 2010-07-15 Cabrera Robert M Immunologic assay for detection of autoantibodies to folate binding protein
US20060115860A1 (en) * 2004-11-26 2006-06-01 Cabrera Robert M Immunologic assay for detection of autoantibodies to folate binding protein
US20080064025A1 (en) * 2005-05-23 2008-03-13 Wei Wen Su Cooperative reporter systems, components, and methods for analyte detection
US7741128B2 (en) 2005-05-23 2010-06-22 University Of Hawaii Cooperative reporter systems, components, and methods for analyte detection
WO2008020823A3 (en) * 2005-05-23 2008-05-29 Univ Hawaii Cooperative reporter systems, components, and methods for analyte detection
WO2008020823A2 (en) * 2005-05-23 2008-02-21 University Of Hawaii Cooperative reporter systems, components, and methods for analyte detection
US20100009342A1 (en) * 2005-09-06 2010-01-14 Life Technologies Corporation Control of chemical modification
US20070154958A1 (en) * 2005-10-13 2007-07-05 Aureon Laboratories, Inc. Multiplex in situ immunohistochemical analysis
US8093012B2 (en) 2005-10-13 2012-01-10 Aureon Laboratories, Inc. Multiplex in situ immunohistochemical analysis
US9645140B2 (en) 2006-02-10 2017-05-09 Life Technologies Corporation Labeling and detection of post translationally modified proteins
US20110207171A1 (en) * 2006-02-10 2011-08-25 Life Technologies Corporation Oligosaccharide modification and labeling of proteins
US10676771B2 (en) 2006-02-10 2020-06-09 Life Technologies Corporation Oligosaccharide modification and labeling of proteins
US20070249014A1 (en) * 2006-02-10 2007-10-25 Invitrogen Corporation Labeling and detection of post translationally modified proteins
US20070190597A1 (en) * 2006-02-10 2007-08-16 Invitrogen Corporation Oligosaccharide modification and labeling of proteins
US8716033B2 (en) 2006-02-10 2014-05-06 Life Technologies Corporation Oligosaccharide modification and labeling of proteins
US8785212B2 (en) 2006-02-10 2014-07-22 Life Technologies Corporation Oligosaccharide modification and labeling of proteins
US8007797B2 (en) 2006-09-28 2011-08-30 Merck Serono S.A. Junctional adhesion molecule-C (JAM-C) binding compounds and methods of their use
US20100034737A1 (en) * 2006-09-28 2010-02-11 Pierre-Yves Dietrich Junctional adhesion molecule-c (jam-c) binding compounds and methods of their use
US7629125B2 (en) 2006-11-16 2009-12-08 General Electric Company Sequential analysis of biological samples
JP2010520989A (en) * 2006-11-16 2010-06-17 ゼネラル・エレクトリック・カンパニイ Continuous analysis of biological samples
US20100120043A1 (en) * 2006-11-16 2010-05-13 General Electric Company Sequential analysis of biological samples
US7741045B2 (en) 2006-11-16 2010-06-22 General Electric Company Sequential analysis of biological samples
US20100047925A1 (en) * 2006-11-16 2010-02-25 General Electric Company Sequential analysis of biological samples
US7741046B2 (en) 2006-11-16 2010-06-22 General Electric Company Sequential analysis of biological samples
US20080118916A1 (en) * 2006-11-16 2008-05-22 General Electric Company Sequential analysis of biological samples
US9201063B2 (en) 2006-11-16 2015-12-01 General Electric Company Sequential analysis of biological samples
US8822147B2 (en) 2006-11-16 2014-09-02 General Electric Company Sequential analysis of biological samples
WO2008133727A2 (en) * 2006-11-16 2008-11-06 General Electric Company Sequential analysis of biological samples with bleaching of intermediate fluorescent signals
WO2008133728A2 (en) * 2006-11-16 2008-11-06 General Electric Company Sequential analysis of biological samples with intermediate bleaching of fluorescence detector
US20080118944A1 (en) * 2006-11-16 2008-05-22 General Electric Company Sequential analysis of biological samples
US20080118934A1 (en) * 2006-11-16 2008-05-22 General Electric Company Sequential analysis of biological samples
WO2008133728A3 (en) * 2006-11-16 2009-01-22 Gen Electric Sequential analysis of biological samples with intermediate bleaching of fluorescence detector
WO2008064067A3 (en) * 2006-11-16 2008-07-10 Gen Electric Method for the sequential analysis of different components in a single biological sample and reagents therefor
WO2008064067A2 (en) * 2006-11-16 2008-05-29 General Electric Company Method for the sequential analysis of different components in a single biological sample and reagents therefor
WO2008133727A3 (en) * 2006-11-16 2009-01-22 Gen Electric Sequential analysis of biological samples with bleaching of intermediate fluorescent signals
WO2008133729A3 (en) * 2006-11-16 2009-01-08 Gen Electric Sequential analysis of biological samples with intermediate bleaching of fluorescence detector
EP2381257A1 (en) * 2006-11-16 2011-10-26 General Electric Company Sequential analysis of biological samples
EP2383574A3 (en) * 2006-11-16 2011-11-30 General Electric Company Sequential analysis of biological samples
US9518982B2 (en) 2006-11-16 2016-12-13 General Electric Company Sequential analysis of biological samples
WO2008133729A2 (en) * 2006-11-16 2008-11-06 General Electric Company Sequential analysis of biological samples with intermediate bleaching of fluorescence detector
US8305579B2 (en) 2006-11-16 2012-11-06 Thomas Pirrie Treynor Sequential analysis of biological samples
US20080233660A1 (en) * 2007-03-23 2008-09-25 Mu Bioteknik Ab Solid phase labeling method
US20080286881A1 (en) * 2007-05-14 2008-11-20 Apel William A Compositions and methods for combining report antibodies
US20090035216A1 (en) * 2007-08-03 2009-02-05 Biomonitor Aps Method for determining in vivo biopharmaceutical concentration or bioavailability
US20100255606A1 (en) * 2007-10-05 2010-10-07 Nobuhiko Sato Labeled peptide having activity of binding to immunoglobulin and/or immunoglobulin complex and method of detecting or assaying immunoglobulin by using the peptide
US20100330592A1 (en) * 2008-01-29 2010-12-30 Key Marc E Method for detecting truncated molecules
US8344115B2 (en) * 2008-04-21 2013-01-01 Chan-Sui Pang, legal representative Immunoassay for specific determination of S-adenosylmethionine and analogs thereof in biological samples
US20090263879A1 (en) * 2008-04-21 2009-10-22 Chiu Chin Chang Immunoassay for specific determination of S-adenosylmethionine and analogs thereof in biological samples
US20100261203A1 (en) * 2009-04-07 2010-10-14 National Institute Of Transplantation Foundation Methods and kits for screening transplant recipients and candidates
US9528988B2 (en) * 2009-04-07 2016-12-27 National Institute Of Transplantation Foundation Methods and kits for screening transplant recipients and candidates
US20110065601A1 (en) * 2009-09-17 2011-03-17 Battelle Energy Alliance, Llc Identification of discriminant proteins through antibody profiling, methods and apparatus for identifying an individual
US20110065594A1 (en) * 2009-09-17 2011-03-17 Battelle Energy Alliance, Llc Identification of discriminant proteins through antibody profiling, methods and apparatus for identifying an individual
US8969009B2 (en) 2009-09-17 2015-03-03 Vicki S. Thompson Identification of discriminant proteins through antibody profiling, methods and apparatus for identifying an individual
US9410965B2 (en) 2009-09-17 2016-08-09 Battelle Energy Alliance, Llc Identification of discriminant proteins through antibody profiling, methods and apparatus for identifying an individual
WO2011048184A1 (en) * 2009-10-21 2011-04-28 General Electric Company Detection of plurality of targets in biological samples
US9677125B2 (en) 2009-10-21 2017-06-13 General Electric Company Detection of plurality of targets in biological samples
US20110092381A1 (en) * 2009-10-21 2011-04-21 General Electric Company Detection of plurality of targets in biological samples
CN102803510A (en) * 2009-10-21 2012-11-28 通用电气公司 Detection of plurality of targets in biological samples
US20130323717A1 (en) * 2009-12-15 2013-12-05 MuHyeon CHOE Repeat-chain for the production of dimer, multimer, multimer complex and super-complex
US10822396B2 (en) * 2009-12-15 2020-11-03 MuHyeon CHOE Repeat-chain for the production of dimer, multimer, multimer complex and super-complex
US20210087254A1 (en) * 2009-12-15 2021-03-25 MuHyeon CHOE Repeat-chain for the production of dimer, multimer, multimer complex and super-complex
US9739773B1 (en) 2010-08-13 2017-08-22 David Gordon Bermudes Compositions and methods for determining successful immunization by one or more vaccines
US10024796B2 (en) 2010-10-29 2018-07-17 President And Fellows Of Harvard College Nucleic acid nanostructure barcode probes
US10876971B2 (en) 2010-10-29 2020-12-29 President And Fellows Of Harvard College Nucleic acid nanostructure barcode probes
JPWO2013035688A1 (en) * 2011-09-09 2015-03-23 コニカミノルタ株式会社 Tissue staining method
US11435348B2 (en) 2011-09-09 2022-09-06 Konica Minolta, Inc. Tissue staining method
US20140212889A1 (en) * 2011-09-09 2014-07-31 Tohoku University Method for staining tissue
US10551378B2 (en) * 2011-09-09 2020-02-04 Konica Minolta, Inc. Tissue staining method
US11906523B2 (en) 2012-03-27 2024-02-20 Ventana Medical Systems, Inc. Signaling conjugates and methods of use
US10557851B2 (en) 2012-03-27 2020-02-11 Ventana Medical Systems, Inc. Signaling conjugates and methods of use
US20150102234A1 (en) * 2012-05-09 2015-04-16 Life Technologies Corporation Systems and method for fluorescence imaging
US11536715B2 (en) 2013-07-30 2022-12-27 President And Fellows Of Harvard College Quantitative DNA-based imaging and super-resolution imaging
US10294510B2 (en) 2014-03-11 2019-05-21 President And Fellows Of Harvard College High-throughput and highly multiplexed imaging with programmable nucleic acid probes
US9944972B2 (en) 2014-03-11 2018-04-17 President And Fellows Of Harvard College High-throughput and highly multiplexed imaging with programmable nucleic acid probes
US10190151B2 (en) 2014-03-11 2019-01-29 President And Fellows Of Harvard College High-throughput and highly multiplexed imaging with programmable nucleic acid probes
WO2015184144A1 (en) * 2014-05-28 2015-12-03 Siemens Healthcare Diagnostics Inc. Rare molecule signal amplification
AT517379B1 (en) * 2015-06-26 2017-03-15 Technische Universität Graz fusion protein
AT517379A1 (en) * 2015-06-26 2017-01-15 Technische Universität Graz fusion protein
US11092606B2 (en) 2015-08-07 2021-08-17 President And Fellows Of Harvard College Super resolution imaging of protein-protein interactions
EP3465162A4 (en) * 2016-06-02 2020-03-25 Ultivue, Inc. Compositions and methods to expedite antibody-based exchange imaging
US11382986B2 (en) * 2016-06-02 2022-07-12 Ultivue, Inc. Compositions and methods to expedite antibody-based exchange imaging
US20190000988A1 (en) * 2016-06-02 2019-01-03 Ultivue, Inc. Compositions and methods to expedite antibody-based exchange imaging
CN109891218A (en) * 2016-06-02 2019-06-14 乌尔蒂维尤股份有限公司 Promote the composition and method of the exchange imaging based on antibody
US11754562B2 (en) 2016-12-09 2023-09-12 Ultivue, Inc. Methods for multiplex imaging using labeled nucleic acid imaging agents
EP3734278A4 (en) * 2017-12-27 2020-12-30 Konica Minolta, Inc. Information acquisition system
WO2021084116A3 (en) * 2018-05-03 2021-06-10 Mursla Limited Biosensor activation and conditioning method
US11738342B2 (en) 2018-05-03 2023-08-29 Mursla Limited Biosensor activation and conditioning method and system
US11865539B2 (en) 2018-05-03 2024-01-09 Mursla Limited Biosensor activation and conditioning method and system
CN114002438A (en) * 2021-11-04 2022-02-01 河南赛诺特生物技术有限公司 Homogeneous immunohistochemical staining method for mouse-anti-mouse tissue
CN116004221A (en) * 2022-12-15 2023-04-25 湖南卓润生物科技有限公司 New application of cyclic peptide, acridine labeled complex, preparation method and detection kit

Also Published As

Publication number Publication date
GB0410515D0 (en) 2004-06-16
GB2397302A (en) 2004-07-21
EP2259067A3 (en) 2011-03-09
EP1442302B1 (en) 2009-07-08
ATE436022T1 (en) 2009-07-15
EP2113773B1 (en) 2012-06-27
WO2003030817A2 (en) 2003-04-17
JP2007183291A (en) 2007-07-19
JP4977512B2 (en) 2012-07-18
EP1442302A2 (en) 2004-08-04
EP2259067B1 (en) 2012-06-27
US8323903B2 (en) 2012-12-04
JP2006513398A (en) 2006-04-20
CA2462280C (en) 2008-03-11
EP1442302A4 (en) 2007-03-28
EP2113773A2 (en) 2009-11-04
GB2397302B (en) 2005-06-08
WO2003030817A3 (en) 2003-09-18
EP2259067A2 (en) 2010-12-08
EP2113773A3 (en) 2010-02-17
WO2003030817B1 (en) 2003-11-06
DE60232904D1 (en) 2009-08-20
CA2462280A1 (en) 2003-04-17
US20070269902A1 (en) 2007-11-22

Similar Documents

Publication Publication Date Title
US8323903B2 (en) Antibody complexes and methods for immunolabeling
US8535894B2 (en) Antibody complexes and methods for immunolabeling
JP6646537B2 (en) Modification and labeling of proteins with oligosaccharides
JP7000489B2 (en) Haptens, hapten conjugates, their compositions and methods of their manufacture and use
US20210215580A1 (en) Combining modified antibodies with expansion microscopy for in-situ, spatially-resolved proteomics
US8093012B2 (en) Multiplex in situ immunohistochemical analysis
US20190112356A1 (en) High-affinity immunopolymers
JP2013056934A6 (en) Method for modifying and labeling proteins with oligosaccharides
JP2013056934A (en) Methods of modifying and labeling proteins with oligosaccharide
JP2009531289A6 (en) Method for modifying and labeling proteins with oligosaccharides
AU2002332013A1 (en) Antibody complexes and methods for immunolabeling

Legal Events

Date Code Title Description
AS Assignment

Owner name: INVITROGEN CORPORATION, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BEECHEM, JOSEPH M;REEL/FRAME:019604/0610

Effective date: 20070724

AS Assignment

Owner name: MOLECULAR PROBES, INC., OREGON

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ARCHER, ROBERT M.;HAGEN, DAVID C.;HAUGLAND, ROSARIA P.;AND OTHERS;REEL/FRAME:019847/0273;SIGNING DATES FROM 20030722 TO 20030819

Owner name: MOLECULAR PROBES, INC., OREGON

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ARCHER, ROBERT M.;HAGEN, DAVID C.;HAUGLAND, ROSARIA P.;AND OTHERS;SIGNING DATES FROM 20030722 TO 20030819;REEL/FRAME:019847/0273

AS Assignment

Owner name: BANK OF AMERICA, N.A., AS COLLATERAL AGENT, WASHIN

Free format text: SECURITY AGREEMENT;ASSIGNOR:LIFE TECHNOLOGIES CORPORATION;REEL/FRAME:021975/0467

Effective date: 20081121

Owner name: BANK OF AMERICA, N.A., AS COLLATERAL AGENT,WASHING

Free format text: SECURITY AGREEMENT;ASSIGNOR:LIFE TECHNOLOGIES CORPORATION;REEL/FRAME:021975/0467

Effective date: 20081121

AS Assignment

Owner name: LIFE TECHNOLOGIES CORPORATION, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MOLECULAR PROBES, INC.;REEL/FRAME:022802/0726

Effective date: 20090529

Owner name: LIFE TECHNOLOGIES CORPORATION,CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MOLECULAR PROBES, INC.;REEL/FRAME:022802/0726

Effective date: 20090529

AS Assignment

Owner name: LIFE TECHNOLOGIES CORPORATION,CALIFORNIA

Free format text: MERGER;ASSIGNOR:INVITROGEN CORPORATION;REEL/FRAME:023882/0551

Effective date: 20081121

Owner name: LIFE TECHNOLOGIES CORPORATION, CALIFORNIA

Free format text: MERGER;ASSIGNOR:INVITROGEN CORPORATION;REEL/FRAME:023882/0551

Effective date: 20081121

STCF Information on status: patent grant

Free format text: PATENTED CASE

AS Assignment

Owner name: LIFE TECHNOLOGIES CORPORATION, CALIFORNIA

Free format text: LIEN RELEASE;ASSIGNOR:BANK OF AMERICA, N.A.;REEL/FRAME:030182/0461

Effective date: 20100528

AS Assignment

Owner name: LIFE TECHNOLOGIES CORPORATION, CALIFORNIA

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE APPLICATION NO 09452626 PREVIOUSLY RECORDED ON REEL 023882 FRAME 0551. ASSIGNOR(S) HEREBY CONFIRMS THE MERGER SHOULD NOT HAVE BEEN RECORDED AGAINST THIS PATENT APPLICATION NUMBER;ASSIGNOR:INVITROGEN CORPORATION;REEL/FRAME:034217/0490

Effective date: 20081121

CC Certificate of correction
FPAY Fee payment

Year of fee payment: 4

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 8